Next Article in Journal
Field-Crop Soils in Eastern France: Coldspots of Azole-Resistant Aspergillus fumigatus
Next Article in Special Issue
Mitogenome Variations in a Global Population of Aspergillus fumigatus
Previous Article in Journal
Phenotypic Array for Identification and Screening of Antifungals against Aspergillus Isolates from Respiratory Infections in KwaZulu Natal, South Africa
Previous Article in Special Issue
Global Analyses of Multi-Locus Sequence Typing Data Reveal Geographic Differentiation, Hybridization, and Recombination in the Cryptococcus gattii Species Complex
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Innate Pulmonary Phagocytes and Their Interactions with Pathogenic Cryptococcus Species

Department of Microbiology and Molecular Genetics, Oklahoma State University, 307 Life Science East, Stillwater, OK 74078, USA
*
Author to whom correspondence should be addressed.
J. Fungi 2023, 9(6), 617; https://doi.org/10.3390/jof9060617
Submission received: 5 May 2023 / Revised: 23 May 2023 / Accepted: 25 May 2023 / Published: 27 May 2023

Abstract

:
Cryptococcus neoformans is an opportunistic fungal pathogen that causes over 180,000 annual deaths in HIV/AIDS patients. Innate phagocytes in the lungs, such as dendritic cells (DCs) and macrophages, are the first cells to interact with the pathogen. Neutrophils, another innate phagocyte, are recruited to the lungs during cryptococcal infection. These innate cells are involved in early detection of C. neoformans, as well as the removal and clearance of cryptococcal infections. However, C. neoformans has developed ways to interfere with these processes, allowing for the evasion of the host’s innate immune system. Additionally, the innate immune cells have the ability to aid in cryptococcal pathogenesis. This review discusses recent literature on the interactions of innate pulmonary phagocytes with C. neoformans.

1. Introduction

Cryptococcus neoformans is an encapsulated fungal pathogen that primarily causes infections in immunocompromised individuals. It is found throughout the environment in soil, decaying wood, and bird excrement [1,2]. Infection by the pathogen occurs via the inhalation of the basidospores or yeast from environmental reservoirs into the pulmonary alveoli, where they initially interact with the resident host innate immune cells [2,3,4]. These cells include subsets of macrophages and dendritic cells (DCs), which are involved in the early recognition as well as removal and clearance of the cryptococcal infection [5,6,7,8,9,10,11,12,13,14]. Following initial infection, neutrophils are recruited to the lung [15,16,17]. Recent studies have found both beneficial and damaging abilities of these innate immune cells during cryptococcal pathogenesis. They have examined the potential of intracellular growth of C. neoformans in macrophages and neutrophils, as well as the use of anti-cryptococcal abilities of DCs in host defense [12,17,18,19,20,21,22,23,24].
In this review, recent literature on the cryptococcal interactions between DCs, macrophages, and neutrophils during infection will be discussed, including a closer look at the potential damaging ability of these cells to the host during cryptococcal pathogenesis.

2. Dendritic Cells

Dendritic cells (DCs) function as one of the first types of immune cells to encounter airway pathogens. DCs are resident cells of the lungs and serve as sentinels of the immune system, recognizing antigens as they are inhaled into the lungs (reviewed in [25]). As phagocytes, DCs have the ability to recognize, engulf, and destroy these pathogens. Additionally, they can circulate to regional lymph nodes for antigen presentation to naïve T cells, activating the adaptive immune response [12,15,26]. Initial recognition of cryptococcal cells by DCs occurs in the lungs. However, cryptococcal cells have a capsule composed of galactoxylomannan (GalXM) and glucuronoxylomannan (GXM). These components have an anti-phagocytic influence on phagocytes, allowing them to evade detection [27,28,29,30,31]. Opsonization by a complement or by anti-capsular monoclonal antibodies negates the anti-phagocytic ability of the polysaccharide capsule, allowing DCs to engulf cryptococcal cells [32,33]. In addition to conventional DCs, plasmacytoid DCs (pDCs) are a rare population of peripheral blood mononuclear cells (PBMCs) found in the blood circulation [34], and these cells also interact with C. neoformans [35].

2.1. Recognition and Activation

Pattern recognition receptors (PRRs) initiate all the fundamental functions of DCs during microbial infections. Found on the cell surface and internally, PRRs are designed to recognize a corresponding set of molecular patterns or microbial pathogen-associated molecular patterns (PAMPs) released by damaged tissues. Association of the PPRs with their PAMPs triggers a cascade of signaling, resulting in the activation of DCs, along with other functions involved in microbial immunity [36,37]. Several studies have examined the role of PRRs during DC recognition and activation by C. neoformans [35,38,39,40,41,42,43,44]. Toll-like receptors (TLRs) are PRRs that are expressed on the surface of cells or on the endosomal membrane. During cryptococcal infections, both TLR2 and TLR4 are active, and with the aid of CD14 and CD11/18, bind to polysaccharides in the cryptococcal capsule [38,40]. The importance of these two receptors for anti-cryptococcal immunity has been debated [38,40,45]. TLR2 was shown in vivo to have a role in the survival of mice during infection with the C. neoformans strain H99 [46]. However, other studies have found that neither TLR2 nor TLR4 are essential for protective responses, despite their ability to recognize components of the cryptococcal capsule [38,39,40]. While TLR2 and TLR4 were not required, the adaptor molecule MyD88 was required, pointing to the involvement of signaling by some other TLR [38]. TLR2−/− and TLR4−/− mice had similar survival rates and expression of TNF-α, interleukin 1β (IL-1β), IL-12p40, and IL-6 compared to wild-type (WT) mice [38,40]. However, numerous studies have identified TLR9 as necessary for host immunity against the fungal pathogen [40,43,46,47,48,49]. TLR9 is capable of activating myeloid DCs through the recognition of URA5 C. neoformans DNA [50,51]. Traditionally, TLR9 is associated with promoting a Th1 protective response and decreasing Th2 hallmarks [48]. Mice deficient in TLR9 were shown to be unable to control cryptococcal infections due to an impairment in cytokine production, which results in an altered adaptive immune response [40,43,49]. This mechanism of impairment is due to a reduction in DC activation in TLR9−/− mice, indicating that TLR9 is important for the activation of DCs and clearance of the cryptococcal cells [38,43,45,46,47,49].
In addition to TLRs, C-type lectin receptors (CLRs) are involved in the recognition of fungal pathogens and recruitment of DCs (reviewed in [52]) [53]. CLRs are molecules with the capability to detect the polysaccharides within the C. neoformans polysaccharide-enriched cell wall and capsule, which is suggested to play a role in the recognition of the fungus by the host’s immune system [53,54]. While the literature has not identified specific CLRs that DCs use for recognition of C. neoformans, Dectin-2 is important in the production of cytokines by DCs in response to cryptococcal infections in vivo and ex vivo [41,42,44,53]. Mice lacking Dectin-2 or the adaptor molecule that induces signaling by CLRs, caspase recruitment domain-containing protein 9 (CARD9), displayed a reduction in C. neoformans uptake by DCs [53]. However, in another study, Dectin-2 was shown to have no effect on the control of the fungal burden by DCs [41]. Interestingly, the signaling molecules SyK and PI3K were shown to have involvement in DC phagocytosis of the fungal cells. When SyK, a molecule involved in activation by CLR, and a PI3K inhibitor were inhibited, there was a reduction in phagocytosis by BMDCs similar to Dectin-2 knockouts and CARD9 knockout mice [55,56]. Additionally, inhibition of the molecule PI3K resulted in complete abrogation of cryptococcal phagocytosis by BMDCs, indicating that it is essential for DCs to phagocytose C. neoformans [55,56]. Moreover, Dectin-3 is required for the recognition and phagocytosis of cryptococcal cells by pDCs, and this is dependent on the host species the cell is derived from. In human pDCs, Dectin-3 is only required for inhibition of cryptococcal growth but is not required for the uptake of cryptococcal cells [35]. Recognition of C. neoformans by DCs is summarized in Figure 1.
After the uptake of cryptococcal cells by DCs through a zipper phagocytosis method, the fungal cell enters the endolysosomal pathway and acquires a lysosomal marker LAMP-1+. There, they are killed through both oxidative and non-oxidative methods [12,32,57,58]. Several in vitro studies examining lysosomal extract have shown it has a direct antifungal activity against C. neoformans [12,18,58]. However, the exact mechanism and lysosomal components involved in the antifungal activity are not completely known. There is evidence that cathepsin B, a component of the lysosomal extract, possesses the ability to kill C. neoformans through the osmotic lysis of cryptococcal cells via damage to their cell wall [58]. Additionally, lysosomal components are being studied in vitro, and six antifungal components (coronin, NOSTRIN, MPO, MMP25, and HNE) have been identified as having antifungal activity in a dose-dependent manner [18]. In addition to conventional DCs, pDCs can also interact with C. neoformans, but little is known about their role in the host’s protective immune response against Cryptococcus. Both murine and human pDCs have been studied and have displayed the ability to inhibit the growth of cryptococcal cells [35]. pDCs have demonstrated anticryptococcal activity by reactive oxygen species (ROS), which is dependent on recognition by Dectin-3 [35].

2.2. Inhibition of DC Maturation

Although they are not the only innate immune cell with the capability to activate T cells, DCs are one of the most proficient, requiring only a select few for the activation of naïve T cells, initiating the adaptive immune response [59]. However, to accomplish this process of DC-mediated T cell activation, the cells must undergo maturation through the increased surface presentation of MHC-II and co-stimulatory molecules (CD80 and CD86) [37,60,61,62]. As mentioned before, GXM has suppressive effects on the host immune response and prevents the phagocytosis of cryptococcal cells, and it non-specifically downregulates T cell proliferation [27]. Furthermore, interactions with cryptococcal mannoproteins can prevent the maturation of DCs in encapsulated strains [28,31,63]. Without proper maturation (increased MHC-II and co-stimulatory molecules), DCs cannot induce a T cell response for proper clearance of the pathogen. With opsonization by a complement or an anti-capsular antibody, DCs are able to recognize C. neoformans through CD32 (FcγRII) and CD16 (FcγRIII) to negate these effects [15,31,59]. However, the presence of the capsule is not the only component involved in inhibition of DC maturation. In three acapsular cryptococcal mutants, it was shown that they cause variable stimulation of DC maturation. The cap10 strain was incapable of inducing DC maturation, while both the cap59 and cap67 strains were able to induce the expression of the surface molecule MHC-II and co-stimulatory markers CD86 and CD80 after uptake [63]. However, the cap59 mutant lost its ability to induce DC maturation after incubation with WT C. neoformans, which suggests that intact GXM is not required for DC maturation to be inhibited [63,64]. This is a result of the association of glucuronoxylomannan present on the WT cells with the cell surface of the acapsular mutant cap59, preventing DC activation [63].

2.3. Pulmonary DC Interactions with C. neoformans

As C. neoformans typically enters through the respiratory tract, the DCs in the lungs are most important in fungal recognition and control. Multiple pulmonary conventional DC subsets in both human and murine lungs have been identified by surface markers [65,66,67,68,69,70,71,72]. Through this process, three subsets of human pulmonary conventional DCs (CD207+, CD14+CD1c+, and CD14CD1c+) and two subsets of murine pulmonary conventional DCs (CD103+ and CD11b+/monocyte-derived DCs or moDCs) have been identified and profiled. All identified human DCs have been identified as possessing the ability to phagocytose and actively kill cryptococcal cells ex vivo. Both subsets of murine conventional DCs have been shown to interact with cryptococcal cells but have different interactions with C. neoformans ex vivo. Neither subset of murine pulmonary DCs was shown to actively kill cryptococcal cells. Interestingly, male CD11b+/moDCs were identified as being capable of enhancing the cellular growth of C. neoformans ex vivo. [73]. Interestingly, the CD11b+/moDCs were shown to infiltrate to the lungs during cryptococcal infection via trafficking by CCR2, and in CCR2-deficient mice, cryptococcal infection led to a shift to Th2 type responses, including increased collagen deposition and increased IL-4, indicating a non-protective immune response [74].
As previously stated, DCs have the capability to uptake and kill cryptococcal cells, which allows for the maturation of the DCs and the activation of the adaptive immune system. However, not all species of Cryptococcus induce DC-mediated T cell activation [37]. Although Cryptococcus gattii is genetically similar to C. neoformans, this organism interacts differently with immune cells. Due to their unique capsule, the Cryptococcus gattii strain R265 has the ability to suppress human DC-mediated T cell activation despite phagocytosis and cryptococcal killing by the DCs [37,75]. After phagocytosing C. gattii R265, the DCs fail to upregulate surface markers such as CD83, CD32, CD86, and MR, which are associated with DC maturation [75].

3. Macrophages

Macrophages are a heterogenous group of immune cells that are either tissue residents or recruited and interact with C. neoformans. They can act either as antifungal cells or they can allow intracellular growth and replication of C. neoformans cells. As we will discuss, these outcomes rely on multiple factors, such as macrophage activation status as well as the macrophage subset.

3.1. Activation

Macrophage polarization is a continual balance of altering the phenotypes with different functions from tissue repair to antimicrobial activity to maintain and protect the human host from invading pathogens [76]. The gene expression profiles of macrophages have a dynamic flexibility that allows them to alter their activation phenotype based on the changes within their environment [77,78,79,80,81,82,83,84]. As a long-living and self-sustainable resident of the lung alveolar airspaces, the ability of macrophages to be adaptable to their environment is critical in providing protection against invading pulmonary pathogens, such as C. neoformans. Otherwise, they would not be able to adjust to various conditions [77,84]. Macrophages can repolarize within hours in response to a different invading pathogen and immune cell-derived signals such as cytokines [82].
Polarization bias of macrophages is influenced by cytokine production, secreted cell byproducts, and extracellular expression of receptors. Macrophage phenotypes are broadly classified as either classical (M1) or alternative (M2), with each type having specific functions during the immune response [81,85,86,87]. In addition, the macrophage phenotype is plastic and can change with the local cytokine microenvironment [82,88]. The capability of a macrophage to control cryptococcal growth during infection is reliant on the predominant type of macrophage activation [17,82,89,90,91,92,93]. Polarization of macrophages between the two phenotypes is associated with a Th1- or Th2-dominated adaptive immune response. During a Th1-type immune response, combined with elevated levels of gamma interferon (IFN-γ), macrophages shift predominately towards an M1 phenotype [86]. M1 macrophages are associated with a reduction in the fungal burden and enhanced fungicidal activity through the production of nitric oxide (NO) and reactive oxygen species (ROS) [82,86,94]. Activation of the STAT1-mediated signaling pathway is required for the production of NO by M1 macrophages. In STAT1 KO mice and STAT1 conditional KO mice infected with IFN-γ-producing C. neoformans, the mice had an enhanced fungal burden, an enhanced M2 macrophage activation, and a reduction in fungicidal activity when compared to WT mice [95,96,97,98]. STAT1-deficient mice favored Arg1 production compared to inducible nitric oxide synthase (iNOS) production (NOS2), an indicator of M2 macrophage activation [97].
While the key initiator of macrophage polarization during C. neoformans infections is based on the fluctuation of Th1 and Th2 cytokines in the microenvironment, interactions with Cryptococcus may also influence the macrophage polarization state [89,99]. In vitro, C. neoformans was shown to suppress NO production by inhibiting NOS2 expression, resulting in the induction of an M2 macrophage-like state [100,101]. NO is one of the effector molecules involved in anti-cryptococcal activity of M1 macrophages [17,20,90,92,93,95,96,97,102,103,104,105,106]. In both iNOS-deficient mice and iNOS-inhibited WT mice, there was an inability to control intracellular growth of C. neoformans within macrophages. This effect remained even in the presence of ROS production [97].
Changes in the gene expression of Cryptococcus-infected macrophages have been extensively studied. However, differences in gene expression are seen between studies. This may potentially be due to the use of various cell lines versus primary cells [19,73,98,107,108,109]. Recent data revealed the ability of C. neoformans to affect the polarization bias of RAW 264.7 macrophages, a murine–leukemia macrophage-like cell line. These cells experienced alterations in genes associated with both lysosomal function and phagocytosis, shifting the polarization of the cells toward a more naïve M0-like state [107]. Additionally, in vivo live C. neoformans cells after uptake by murine alveolar macrophages could cause impairment of the lysosome, which can promote an increase in cell proliferation within the phagocyte [88]. During C. neoformans infections, the lysosomes of these cells were shown to have fragmented phagolysosomal membranes that become permeable to macromolecules [110,111]. C. neoformans can also lead to production of actin flashes outside the phagosome, which presumably prevents the organism from being expulsed from the macrophage [112]. Cryptococcus-induced permeabilization of the phagolysosomal membrane is an indicator of lysosomal damage and results in the leakage of phagolysosomal contents into the cytosol of the immune cell [110,111,113]. This leakage has previously been associated with the induction of apoptosis, as well as a loss of antimicrobial contents of the phagolysosome [114]. However, this effect was shown to be negated by the introduction of IFN-γ to the macrophages during their interaction with the cryptococcal cells, allowing the immune cells to maintain fungicidal activity to C. neoformans [88,91]. This suggests that an intact phagolysosomal membrane is vital in determining the outcome of a Cryptococcus and pulmonary macrophage interaction [88,113].

3.2. Trafficking of C. neoformans

In contrast to M1 macrophages, M2 macrophages are associated with being the primary host cell involved in intracellular growth of C. neoformans [90,93,106,115]. After uptake, cryptococci traffic in the phagosome, which fuses with the lysosome to become a phagolysosome, results in their exposure to NO, ROS, degradative enzymes, and an acidic environment [116,117]. C. neoformans are shown to be capable of hindering acidification of the phagosome, allowing for intracellular proliferation in both human monocyte-derived macrophages and in the J774A.1 macrophage cell line [118]. However, C. neoformans can also survive in an acidic environment [117]. Additional cryptococcal proteins are required for intracellular growth to occur. Replication within macrophages requires the presence of phospholipase B (PLB1), a known factor of Cryptococcus virulence. Deletion of the gene PLB1 in C. neoformans led to reduced survival and replication in macrophages [119,120]. Additionally, F-box protein 1 (fb1) and its substrate inositol phosphosphingolipid-phospholipase C1 are necessary for the spread of the pathogen to the central nervous system (CNS) and for resistance to NO [121]. Following M1 polarization by stimulation of IFN-γ, the fungal pathogens’ ability to induce lysosomal damage is negated and the macrophages exhibit an increase in fungal killing [88].
In addition to intracellular growth, C. neoformans cells are capable of non-lytic exocytosis from macrophages through the escape of cryptococci from the phagocyte without destroying the immune cell in the process (in vitro and in vivo models [7,122,123]). Both cryptococcal virulence factors and host factors regulate non-lytic exocytosis of the cells [123,124]. The capability of the fungal pathogen to successfully survive and replicate within the host immune cells contributes to increased fungal dissemination within the host. It has been suggested that the macrophages act as a “Trojan Horse” during cryptococcal pathogenesis, carrying the pathogen across the blood–brain barrier (BBB) by trans-endothelial pores [125,126]. Once the fungal organism crosses the BBB, it can lead to the development of a CNS infection and meningoencephalitis [126,127,128,129]. Unfortunately, this infection is extremely dangerous, with survivors often developing adverse effects including neurological deficits [130,131]. After invasion of the CNS, the microglial response is essential against the invading C. neoformans. Microglia are brain-resident macrophages and are found throughout the parenchyma of the brain. Similar to other innate immune cells, they possess two states. In a healthy host, they remain in a resting state until activation [132]. Upon activation, they change their overall shape into an amoeboid-like morphology, which allows them to phagocytose pathogens, infected cells, and dead neurons [133,134,135,136,137,138]. These cells can recognize fungal PAMPs via TLRs. Recognition of fungal PAMPs promotes the release of antimicrobial molecules and proinflammatory cytokines into the microenvironment for the recruitment of innate and adaptive immune cells [5,139]. Interestingly, there are differences between murine and human microglia, with human cells being unable to kill C. neoformans [140]. Since the secretion of NO is positively correlated with the cells’ ability to kill C. neoformans, it is potentially due to the inability of human microglia to produce a sufficient amount of NO compared to the murine microglia [140,141,142].
Dissemination to other organs, such as the liver, can also occur during cryptococcosis. However, macrophages resident to the liver, Kupffer cells (KCs), are able to modulate liver infection by Cryptococcus [143]. Phagocytosis by KC involves complement receptors CR3 and CRIg and scavenger receptors, and inhibition of Cryptococcus growth is dependent on IFN-γ but not on IFNγR signaling [143].
Ly6c+-expressing inflammatory monocytes (IFM) are precursors to both macrophages and DCs and, like their derivatives, they are able to inhibit the growth of cryptococcal cells [74,144,145,146,147,148,149,150]. However, despite their ability to inhibit cryptococcal growth, they may also be involved in the progression of C. neoformans infections. When IFMs were ablated in vivo, there was an improvement in the fungal burden. A transcriptional analysis of the IFMs identified M2 surface marker expression when challenged by C. neoformans [144]. M2 macrophages are associated with host tissue repair and homeostasis. They are not typically associated with inflammatory-type cells [151]. However, the M2 macrophage phenotype is shown to be involved in the intracellular growth of C. neoformans [90,93,106,115].

3.3. Pulmonary Macrophage Interactions with C. neoformans

Since C. neoformans is typically inhaled, it is important to investigate the role of pulmonary macrophages. The pulmonary macrophages consist of a diverse population of cells [65,152,153,154]. Originally, it was assumed in the pulmonary region that macrophage populations were divided into alveolar macrophages (AM) and interstitial macrophages (IM). Each was named for their respective designated region of the pulmonary airways and tissues [155]. More recently, however, multiple pulmonary macrophage subsets in both human and murine lungs have been identified by flow cytometry [65,73,108], and their lineages have been defined by lineage tracing [66,68,156,157,158,159,160]. Through this process, three subsets of human pulmonary macrophages (AM, CD14+CD1c, and CD14CD1c) have been identified and profiled in healthy human lungs [65]. All human subsets internalize cryptococci, although AMs and CD14+ macrophages are more efficient at fungal uptake compared to CD14 macrophages [108]. Furthermore, following interactions with Cryptococcus, the fate of the pathogen varied between subsets [108]. While AMs displayed consistent antifungal activity against the fungal pathogen, CD14 and CD14+ macrophages were unable to kill cryptococcal cells after uptake [108]. Transcriptional analyses of these subsets following interaction with C. neoformans compared to the subsets alone revealed changes in gene expression within metabolism (MTRNR2L12, MT-ND6, MT-ATP8, MT-CO3, and MT-CYB) and antigen presentation genes (HLA-A, HLA-B, HLA-C, and HLA-DRA) [108]. Additionally, four murine macrophage subsets (AM, IM, Ly6c+, and Ly6c monocyte-like macrophages) have been identified through flow cytometry [73], as well as gene expression profiling and lineage tracing [66,67,68,70,72,157,159,160,161,162]. All identified murine macrophage subsets can interact with cryptococcal cells, but only female Ly6c- monocyte-like macrophages can significantly inhibit the cellular growth of cryptococcal cells. These cells expressed a significant upregulation in MHC-I and significant regulation in several metabolic genes [73]. The MHC-I antigen presentation pathway is associated with the initiation of the adaptive immune response to virally infected cells, but can also be involved in cross-presentation of cryptococcal antigens [163,164,165].

4. Neutrophils

Neutrophils or polymorphonuclear leukocytes (PMNs) are phagocytes that are recruited during cryptococcal infections to the lungs [9,166,167,168]. They are one of the most abundant types of immune cells present in the human bloodstream and stem from the bone marrow in large amounts of ~1011 cells per day [169]. These cells are essential in the killing and regulation of cryptococcal cells during the initial infection and are shown to have antifungal abilities greater than monocytes and macrophages [115,170,171]. After infection, circulating neutrophils migrate to infection sites and aid in fungal clearance. They can kill using both intracellular and extracellular methods, and by oxidative and non-oxidative mechanisms [9]. Neutrophil swarming is essential for the innate immune cells to accumulate at sites of infection for the clearance of pathogens [172]. In vivo, this process of neutrophil migration is mediated by the presence of complement C3 and C5a-C5aR complement pathways and actin polymerization to the fungal infection site [173,174]. Complement C3 is required for neutrophil swarming to cryptococcal cells. In C3−/− murine models, no interaction between C. neoformans and neutrophils occurred despite the presence of the cryptococcal polysaccharide capsule [174]. This is likely due to a lack of complement C3b opsonization of the fungal cells [175].
C5a-C5aR is important in fungal clearance in three ways. First, C5aR significantly increased the distance they can travel [176]. Secondly, C5aR also enhances the expression of the surface marker CD11b. CD11b is a part of the integrin Mac-1 molecule, a monocyte, neutrophil, and macrophage surface receptor, and along with CD18, forms complement receptor 3 [177,178]. CD11b helps with the adhesion of these phagocytes to various cells and regulates antimicrobial responses, including phagocytosis and migration [178]. Mac-1 binds to its ligand ICAM-1 during cryptococcal infections. Blockage of CD11b almost completely abolishes neutrophil ability to kill cryptococcal cells by preventing the cells from phagocytosing the fungus [176]. Additionally, CD11b is critical for neutrophil transmigration through endothelial cells. CD11b-knockout neutrophils were shown to be defective in intravascular crawling, and the majority failed to go through transendothelial migration [179,180]. Lastly, C5aR signaling mediates the migration of neutrophils to C. neoformans. Interestingly, C5a is not completely required for the swarming of neutrophils around C. neoformans cells [173,176]. While C5−/− mice were shown to have fewer neutrophil clusters compared to WT mice, there was still a presence of neutrophil clustering around C. neoformans. Despite the lack of C5a-C5aR signaling, C. neoformans are still able to be opsonized by C3b/iC3b [175]. However, C5a is required for the ideal killing of the fungus by neutrophils via the formation of a concentration gradient around encapsulated cryptococcal cells [176]. Further studies have showed that the mitogen-activated protein kinase (MAPK) pathway is involved in the migration of neutrophils using the C5a-C5aR complement pathway. Inhibition of the p38 kinase resulted in a decrease in neutrophil-mediated cryptococcal killing and migration [173,181,182,183].
During an infection, neutrophils in close proximity to the infection site are initially recruited via C5a-C5aR signaling as discussed above [184]. Once this occurs, chemotactic neutrophils will secrete leukotriene B4 (LTB4), creating a second chemical gradient that increases the range of the primary chemoattractant [175,185]. When LTB4 synthesis is inhibited in vivo, there is no formation of large neutrophil clusters and there is a significant reduction in neutrophil migration to the lungs, indicating that LTB4 is not only required for the huge migration of distant neutrophils but also for the swarming of neutrophils within the lungs [175,186].
C. neoformans can also influence the neutrophil fungicidal response. Neutrophils can combat invading pathogens by releasing a neutrophil extracellular trap (NET), made up of condensed chromatin with cytosolic and granular proteins [187,188]. These NETs retain and kill the microbe, while protecting the host cells. C. neoformans can inhibit NET production with a component of the capsule, glucuronoxylomannan (GXM) [187]. Cryptococcal capsular components can also inhibit the migration of neutrophils to the infection site. They prevent neutrophil migration by releasing components of their capsule, which interferes with trafficking due to chemokine gradients and neutrophil rolling [189].
Circulating neutrophils are capable of interaction with C. neoformans cells arrested to walls of blood vessels. After engulfment of the adhered cells, they can enter back into the bloodstream with the fungal cell, resulting in the removal of cryptococcal cells from the brain vasculature [23,176,190]. Clearance of fungal cells within the brain vasculature is mainly facilitated by the neutrophils. However, the efficiency of neutrophils in the intravascular clearance of C. neoformans in the brain is significantly lower than in the pulmonary region of the human body [176].
Despite neutrophils being capable of removing and killing C. neoformans, their role in cryptococcal infections is still largely unknown. In vivo studies have presented controversial results, portraying neutrophils as having either protective or damaging roles [115,191]. During the protective immune response, depletion of neutrophils does not affect the fungal burden in pulmonary tissue of mice [16,192], and neutropenic mice survived longer after the initiation of a pulmonary C. neoformans infection than those with normal neutrophil counts [17,193]. In HIV patients with cryptococcal meningitis, an increased neutrophil count was associated with a higher mortality rate [193]. This suggests that neutrophils are not required for the clearance of C. neoformans by the host’s immune system and may actually aid in cryptococcal pathogenesis [16,17,192]. However, the detrimental role of neutrophils during cryptococcal infections is still under debate as the depletion of neutrophils has also been associated with a reduction in fungal clearance in the brain [23].
C. neoformans is a known facultative intracellular pathogen of monocyte-derived macrophages and dendritic cells [110,111,118,194]. However, little is known about the ability of C. neoformans to survive within neutrophils after phagocytosis [23]. Non-lytic exocytosis of C. neoformans has previously been seen in macrophages and monocytes and has long been suspected to contribute to the spread of fungal cells in the neural tissue of the host, resulting in cryptococcal meningitis [123,195,196,197]. However, these innate immune cells are not the only cells involved in the potential trafficking of C. neoformans across the blood–brain barrier. Neutrophils, despite their vital role in the innate immune response, have demonstrated similar behaviors [24]. Neutrophils were shown to both phagocytose and traffic cryptococcal cells to the brain and deposit them into the brain vascular tissue by exocytosis. This may contribute to brain infections and may explain why the fungal burden in the brain decreased in correlation with a reduction in neutrophils present in the blood [23].

5. Conclusions

Innate immune cells are the first to interact with invading pathogens. They aid in preventing the establishment of infections and their potential dissemination from the primary site of infection. While innate immune phagocytes are known to have important roles in the identification and removal of pathogens such as C. neoformans, they can also play a role in cryptococcal pathogenesis. As summarized in Figure 2, clearance of the pathogen by these cells is not always the outcome of the innate immune phagocyte interaction. Recent studies have found both beneficial and damaging abilities of these innate immune cells during cryptococcal pathogenesis. C. neoformans has mechanisms that can interfere with the ability of host phagocytes to recognize, phagocytose, and clear fungi during infection, allowing for the dissemination of the organism to the CNS. Though mechanisms of host evasion by the fungal pathogen have been extensively studied over the years, there is still more to learn, specifically regarding the ability of the microorganism to use the host’s innate immune cells as transportation to extra-pulmonary regions of the human body.

Author Contributions

Conceptualization, B.N.C. and K.L.W.; writing—original draft preparation, B.N.C.; writing—review and editing, K.L.W. and B.N.C.; original figure preparation—B.N.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a research grant, 1P20GM134973-01 (KLW), from the National Institute of General Medical Sciences (GMS) of the National Institutes of Health (NIH) and the G-RISE Fellowship: NIH T32 GM140953 from GMS of the NIH (BNC). The funders had no role in the decision to publish or preparation of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kwon-Chung, K.J.; Fraser, J.A.; Doering, T.L.; Wang, Z.; Janbon, G.; Idnurm, A.; Bahn, Y.S. Cryptococcus neoformans and Cryptococcus gattii, the Etiologic Agents of Cryptococcosis. Cold Spring Harb. Perspect. Med. 2014, 4, a019760. [Google Scholar] [CrossRef]
  2. Perfect, J.R.; Casadevall, A. Cryptococcosis. Infect. Dis. Clin. 2002, 16, 837–874. [Google Scholar] [CrossRef]
  3. Ellis, D.H.; Pfeiffer, T.J. Ecology, life cycle, and infectious propagule of Cryptococcus neoformans. Lancet 1990, 336, 923–925. [Google Scholar] [CrossRef]
  4. Maziarz, E.K.; Perfect, J.R. Cryptococcosis. Infect. Dis. Clin. N. Am. 2016, 30, 179–206. [Google Scholar] [CrossRef]
  5. Redlich, S.; Ribes, S.; Schütze, S.; Eiffert, H.; Nau, R. Toll-like receptor stimulation increases phagocytosis of Cryptococcus neoformans by microglial cells. J. Neuroinflamm. 2013, 10, 841–847. [Google Scholar] [CrossRef] [PubMed]
  6. Chrisman, C.J.; Albuquerque, P.; Guimaraes, A.J.; Nieves, E.; Casadevall, A. Phospholipids Trigger Cryptococcus neoformans Capsular Enlargement during Interactions with Amoebae and Macrophages. PLoS Pathog. 2011, 7, e1002047. [Google Scholar] [CrossRef] [PubMed]
  7. Alvarez, M.; Casadevall, A. Cell-to-cell spread and massive vacuole formation after Cryptococcus neoformans infection of murine macrophages. BMC Immunol. 2007, 8, 16. [Google Scholar] [CrossRef]
  8. Rohatgi, S.; Pirofski, L.-A. Host immunity to Cryptococcus neoformans. Futur. Microbiol. 2015, 10, 565–581. [Google Scholar] [CrossRef] [PubMed]
  9. Mambula, S.S.; Simons, E.R.; Hastey, R.; Selsted, M.E.; Levitz, S.M. Human Neutrophil-Mediated Nonoxidative Antifungal Activity against Cryptococcus neoformans. Infect. Immun. 2000, 68, 6257–6264. [Google Scholar] [CrossRef]
  10. Winterbourn, C.C.; Vissers, M.C.; Kettle, A.J. Myeloperoxidase. Curr. Opin. Hematol. 2000, 7, 53–58. [Google Scholar] [CrossRef]
  11. Alcouloumre, M.S.; Ghannoum, M.A.; Ibrahim, A.S.; Selsted, M.E.; Edwards, J.E., Jr. Fungicidal properties of defensin NP-1 and activity against Cryptococcus neoformans in vitro. Antimicrob. Agents Chemother. 1993, 37, 2628–2632. [Google Scholar] [CrossRef] [PubMed]
  12. Wozniak, K.L.; Levitz, S.M. Cryptococcus neoformans Enters the Endolysosomal Pathway of Dendritic Cells and Is Killed by Lysosomal Components. Infect. Immun. 2008, 76, 4764–4771. [Google Scholar] [CrossRef] [PubMed]
  13. Islam, A.; Li, S.S.; Oykhman, P.; Timm-McCann, M.; Huston, S.M.; Stack, D.; Xiang, R.F.; Kelly, M.M.; Mody, C.H. An Acidic Microenvironment Increases NK Cell Killing of Cryptococcus neoformans and Cryptococcus gattii by Enhancing Perforin Degranulation. PLoS Pathog. 2013, 9, e1003439. [Google Scholar] [CrossRef]
  14. Marr, K.J.; Jones, G.J.; Zheng, C.; Huston, S.M.; Timm-McCann, M.; Islam, A.; Berenger, B.M.; Ma, L.L.; Wiseman, J.C.; Mody, C.H. Cryptococcus neoformans Directly Stimulates Perforin Production and Rearms NK Cells for Enhanced Anticryptococcal Microbicidal Activity. Infect. Immun. 2009, 77, 2436–2446. [Google Scholar] [CrossRef]
  15. 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]
  16. 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 gamma/delta T cells. BMC Immunol. 2012, 13, 65. [Google Scholar] [CrossRef] [PubMed]
  17. 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]
  18. Nelson, B.N.; Beakley, S.G.; Posey, S.; Conn, B.; Maritz, E.; Seshu, J.; Wozniak, K.L. Antifungal activity of dendritic cell lysosomal proteins against Cryptococcus neoformans. Sci. Rep. 2021, 11, 13619. [Google Scholar] [CrossRef]
  19. Coelho, C.; Bocca, A.L.; Casadevall, A. The Intracellular Life of Cryptococcus neoformans. Annu. Rev. Pathol. 2014, 9, 219–238. [Google Scholar] [CrossRef] [PubMed]
  20. 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]
  21. Mansour, M.K.; Reedy, J.L.; Tam, J.M.; Vyas, J.M. Macrophage–Cryptococcus Interactions: An Update. Curr. Fungal Infect. Rep. 2014, 8, 109–115. [Google Scholar] [CrossRef]
  22. Heung, L.J. Innate Immune Responses to Cryptococcus. J. Fungi 2017, 3, 35. [Google Scholar] [CrossRef]
  23. 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]
  24. Yang, X.; Wang, H.; Hu, F.; Chen, X.; Zhang, M. Nonlytic exocytosis of Cryptococcus neoformans from neutrophils in the brain vasculature. Cell Commun. Signal 2019, 17, 117. [Google Scholar] [CrossRef] [PubMed]
  25. Wozniak, K.L. Interactions of Cryptococcus with Dendritic Cells. J. Fungi 2018, 4, 36. [Google Scholar] [CrossRef] [PubMed]
  26. 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]
  27. Yauch, L.E.; Lam, J.S.; Levitz, S.M. Direct Inhibition of T-Cell Responses by the Cryptococcus Capsular Polysaccharide Glucuronoxylomannan. PLoS Pathog. 2006, 2, e120. [Google Scholar] [CrossRef]
  28. Zaragoza, O.; Rodrigues, M.L.; De Jesus, M.; Frases, S.; Dadachova, E.; Casadevall, A. The capsule of the fungal pathogen Cryptococcus neoformans. Adv. Appl. Microbiol. 2009, 68, 133–216. [Google Scholar] [CrossRef] [PubMed]
  29. 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] [CrossRef]
  30. Kozel, T.R.; Pfrommer, G.S.; Guerlain, A.S.; Highison, B.A.; Highison, G.J. Role of the capsule in phagocytosis of Cryptococcus neoformans. Rev. Infect. Dis. 1988, 10 (Suppl. 2), S436–S439. [Google Scholar] [CrossRef]
  31. Vecchiarelli, A.; Pericolini, E.; Gabrielli, E.; Kenno, S.; Perito, S.; Cenci, E.; Monari, C. Elucidating the immunological function of the Cryptococcus neoformans capsule. Futur. Microbiol. 2013, 8, 1107–1116. [Google Scholar] [CrossRef]
  32. 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]
  33. Mukherjee, J.; Scharff, M.D.; Casadevall, A. Protective murine monoclonal antibodies to Cryptococcus neoformans. Infect. Immun. 1992, 60, 4534–4541. [Google Scholar] [CrossRef]
  34. Schmidt, B.; Fujimura, S.H.; Martin, J.N.; Levy, J.A. Variations in Plasmacytoid Dendritic Cell (PDC) and Myeloid Dendritic Cell (MDC) Levels in HIV-Infected Subjects on and off Antiretroviral Therapy. J. Clin. Immunol. 2006, 26, 55–64. [Google Scholar] [CrossRef]
  35. 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]
  36. Olszewski, M.A.; Zhang, Y.; Huffnagle, G.B. Mechanisms of cryptococcal virulence and persistence. Futur. Microbiol. 2010, 5, 1269–1288. [Google Scholar] [CrossRef] [PubMed]
  37. Huston, S.M.; Li, S.S.; Stack, D.; Timm-McCann, M.; Jones, G.J.; Islam, A.; Berenger, B.M.; Xiang, R.F.; Colarusso, P.; Mody, C.H. Cryptococcus gattii Is Killed by Dendritic Cells, but Evades Adaptive Immunity by Failing To Induce Dendritic Cell Maturation. J. Immunol. 2013, 191, 249–261. [Google Scholar] [CrossRef]
  38. 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]
  39. 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]
  40. 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]
  41. 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]
  42. 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]
  43. 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]
  44. 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]
  45. Eastman, A.J.; Osterholzer, J.J.; Olszewski, M.A. Role of dendritic cell–pathogen interactions in the immune response to pulmonary cryptococcal infection. Futur. Microbiol. 2015, 10, 1837–1857. [Google Scholar] [CrossRef]
  46. 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]
  47. Qiu, Y.; Zeltzer, S.; Zhang, Y.; Wang, F.; Chen, G.-H.; Dayrit, J.; Murdock, B.J.; Bhan, U.; Toews, G.B.; Osterholzer, J.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]
  48. 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]
  49. 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]
  50. 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]
  51. 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]
  52. Hardison, S.E.; Brown, G.D. C-type lectin receptors orchestrate antifungal immunity. Nat. Immunol. 2012, 13, 817–822. [Google Scholar] [CrossRef]
  53. Kitai, Y.; Sato, K.; Tanno, D.; Yuan, X.; Umeki, A.; Kasamatsu, J.; Kanno, E.; Tanno, H.; Hara, H.; Yamasaki, S.; et al. Role of Dectin-2 in the Phagocytosis of Cryptococcus neoformans by Dendritic Cells. Infect. Immun. 2021, 89, e00330-21. [Google Scholar] [CrossRef] [PubMed]
  54. Willment, J.A.; Brown, G.D. C-type lectin receptors in antifungal immunity. Trends Microbiol. 2008, 16, 27–32. [Google Scholar] [CrossRef] [PubMed]
  55. Robinson, M.J.; Osorio, F.; Rosas, M.; Freitas, R.P.; Schweighoffer, E.; Groß, O.; Verbeek, J.S.; Ruland, J.; Tybulewicz, V.; Brown, G.D.; et al. Dectin-2 is a Syk-coupled pattern recognition receptor crucial for Th17 responses to fungal infection. J. Exp. Med. 2009, 206, 2037–2051. [Google Scholar] [CrossRef]
  56. Lee, M.J.; Yoshimoto, E.; Saijo, S.; Iwakura, Y.; Lin, X.; Katz, H.R.; Kanaoka, Y.; Barrett, N.A. Phosphoinositide 3-Kinase δ Regulates Dectin-2 Signaling and the Generation of Th2 and Th17 Immunity. J. Immunol. 2016, 197, 278–287. [Google Scholar] [CrossRef]
  57. Artavanis-Tsakonas, K.; Love, J.C.; Ploegh, H.L.; Vyas, J.M. Recruitment of CD63 to Cryptococcus neoformans phagosomes requires acidification. Proc. Natl. Acad. Sci. USA 2006, 103, 15945–15950. [Google Scholar] [CrossRef]
  58. Hole, C.R.; Bui, H.; Wormley, F.L., Jr.; Wozniak, K.L. Mechanisms of Dendritic Cell Lysosomal Killing of Cryptococcus. Sci. Rep. 2012, 2, 739. [Google Scholar] [CrossRef] [PubMed]
  59. Syme, R.M.; Spurrell, J.C.L.; Amankwah, E.K.; Green, F.H.Y.; Mody, C.H. Primary dendritic cells phagocytose Cryptococcus neoformans via mannose receptors and Fcgamma receptor II for presentation to T lymphocytes. Infect. Immun. 2002, 70, 5972–5981. [Google Scholar] [CrossRef]
  60. Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef] [PubMed]
  61. Vecchiarelli, A.; Pietrella, D.; Lupo, P.; Bistoni, F.; McFadden, D.C.; Casadevall, A. The polysaccharide capsule of Cryptococcus neoformans interferes with human dendritic cell maturation and activation. J. Leukoc. Biol. 2003, 74, 370–378. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, A.W.; Truong, T.; Bickham, K.; Fonteneau, J.-F.; Larsson, M.; Da Silva, I.; Somersan, S.; Thomas, E.K.; Bhardwaj, N. A clinical grade cocktail of cytokines and PGE2 results in uniform maturation of human monocyte-derived dendritic cells: Implications for immunotherapy. Vaccine 2002, 20 (Suppl. S4), A8–A22. [Google Scholar] [CrossRef]
  63. Grijpstra, J.; Tefsen, B.; van Die, I.; de Cock, H. The Cryptococcus neoformans cap10 and cap59 mutant strains, affected in glucuronoxylomannan synthesis, differentially activate human dendritic cells. FEMS Immunol. Med. Microbiol. 2009, 57, 142–150. [Google Scholar] [CrossRef]
  64. Nelson, B.N.; Hawkins, A.N.; Wozniak, K.L. Pulmonary Macrophage and Dendritic Cell Responses to Cryptococcus neoformans. Front. Cell. Infect. Microbiol. 2020, 10, 37. [Google Scholar] [CrossRef]
  65. Patel, V.I.; Booth, J.L.; Duggan, E.S.; Cate, S.; White, V.L.; Hutchings, D.; Kovats, S.; Burian, D.M.; Dozmorov, M.; Metcalf, J.P. Transcriptional Classification and Functional Characterization of Human Airway Macrophage and Dendritic Cell Subsets. J. Immunol. 2017, 198, 1183–1201. [Google Scholar] [CrossRef] [PubMed]
  66. Misharin, A.V.; Morales-Nebreda, L.; Mutlu, G.M.; Budinger, G.R.; Perlman, H. Flow Cytometric Analysis of Macrophages and Dendritic Cell Subsets in the Mouse Lung. Am. J. Respir. Cell Mol. Biol. 2013, 49, 503–510. [Google Scholar] [CrossRef] [PubMed]
  67. Zaynagetdinov, R.; Sherrill, T.P.; Kendall, P.L.; Segal, B.H.; Weller, K.P.; Tighe, R.M.; Blackwell, T.S. Identification of Myeloid Cell Subsets in Murine Lungs Using Flow Cytometry. Am. J. Respir. Cell Mol. Biol. 2013, 49, 180–189. [Google Scholar] [CrossRef]
  68. Kopf, M.; Schneider, C.; Nobs, S.P. The development and function of lung-resident macrophages and dendritic cells. Nat. Immunol. 2014, 16, 36–44. [Google Scholar] [CrossRef]
  69. Satpathy, A.T.; Wu, X.; Albring, J.C.; Murphy, K.M. Re(de)fining the dendritic cell lineage. Nat. Immunol. 2012, 13, 1145–1154. [Google Scholar] [CrossRef]
  70. Vermaelen, K.; Pauwels, R. Accurate and simple discrimination of mouse pulmonary dendritic cell and macrophage populations by flow cytometry: Methodology and new insights. Cytometry A 2004, 61, 170–177. [Google Scholar] [CrossRef]
  71. Condon, T.V.; Sawyer, R.T.; Fenton, M.J.; Riches, D.W.H. Lung dendritic cells at the innate-adaptive immune interface. J. Leukoc. Biol. 2011, 90, 883–895. [Google Scholar] [CrossRef] [PubMed]
  72. Shortman, K.; Liu, Y.-J. Mouse and human dendritic cell subtypes. Nat. Rev. Immunol. 2002, 2, 151–161. [Google Scholar] [CrossRef]
  73. Hawkins, A.N.; Determann, B.F., 2nd; Nelson, B.N.; Wozniak, K.L. Transcriptional Changes in Pulmonary Phagocyte Subsets Dictate the Outcome Following Interaction with the Fungal Pathogen Cryptococcus neoformans. Front. Immunol. 2021, 12, 722500. [Google Scholar] [CrossRef] [PubMed]
  74. 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]
  75. Huston, S.M.; Ngamskulrungroj, P.; Xiang, R.F.; Ogbomo, H.; Stack, D.; Li, S.S.; Timm-McCann, M.; Kyei, S.K.; Oykhman, P.; Kwon-Chung, K.J.; et al. Cryptococcus gattii Capsule Blocks Surface Recognition Required for Dendritic Cell Maturation Independent of Internalization and Antigen Processing. J. Immunol. 2016, 196, 1259–1271. [Google Scholar] [CrossRef]
  76. Jablonski, K.A.; Amici, S.A.; Webb, L.M.; Ruiz-Rosado Jde, D.; Popovich, P.G.; Partida-Sanchez, S.; Guerau-de-Arellano, M. Novel Markers to Delineate Murine M1 and M2 Macrophages. PLoS ONE 2015, 10, e0145342. [Google Scholar] [CrossRef] [PubMed]
  77. Fels, A.O.; Cohn, Z.A. The alveolar macrophage. J. Appl. Physiol. 1986, 60, 353–369. [Google Scholar] [CrossRef]
  78. Porcheray, F.; Viaud, S.; Rimaniol, A.C.; Leone, C.; Samah, B.; Dereuddre-Bosquet, N.; Dormont, D.; Gras, G. Macrophage activation switching: An asset for the resolution of inflammation. Clin. Exp. Immunol. 2005, 142, 481–489. [Google Scholar] [CrossRef]
  79. Stout, R.D.; Jiang, C.; Matta, B.; Tietzel, I.; Watkins, S.K.; Suttles, J. Macrophages Sequentially Change Their Functional Phenotype in Response to Changes in Microenvironmental Influences. J. Immunol. 2005, 175, 342–349. [Google Scholar] [CrossRef]
  80. Gratchev, A.; Kzhyshkowska, J.; Köthe, K.; Muller-Molinet, I.; Kannookadan, S.; Utikal, J.; Goerdt, S. Mφ1 and Mφ2 can be re-polarized by Th2 or Th1 cytokines, respectively, and respond to exogenous danger signals. Immunobiology 2006, 211, 473–486. [Google Scholar] [CrossRef] [PubMed]
  81. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef] [PubMed]
  82. 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, e00264-13. [Google Scholar] [CrossRef] [PubMed]
  83. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef]
  84. Xu-Vanpala, S.; Deerhake, M.E.; Wheaton, J.D.; Parker, M.E.; Juvvadi, P.R.; MacIver, N.; Ciofani, M.; Shinohara, M.L. Functional heterogeneity of alveolar macrophage population based on expression of CXCL2. Sci. Immunol. 2020, 5, aba7350. [Google Scholar] [CrossRef]
  85. Stout, R.D.; Suttles, J. Functional plasticity of macrophages: Reversible adaptation to changing microenvironments. J. Leukoc. Biol. 2004, 76, 509–513. [Google Scholar] [CrossRef] [PubMed]
  86. Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef]
  87. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.-A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
  88. Davis, M.J.; Eastman, A.J.; Qiu, Y.; Gregorka, B.; Kozel, T.R.; Osterholzer, J.J.; Curtis, J.L.; Swanson, J.A.; Olszewski, M.A. Cryptococcus neoformans—Induced Macrophage Lysosome Damage Crucially Contributes to Fungal Virulence. J. Immunol. 2015, 194, 2219–2231. [Google Scholar] [CrossRef]
  89. 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]
  90. Müller, U.; Stenzel, W.; Köhler, G.; Werner, C.; Polte, T.; Hansen, G.; Schütze, N.; Straubinger, R.K.; Blessing, M.; McKenzie, A.N.J.; et al. IL-13 Induces Disease-Promoting Type 2 Cytokines, Alternatively Activated Macrophages and Allergic Inflammation during Pulmonary Infection of Mice with Cryptococcus neoformans. J. Immunol. 2007, 179, 5367–5377. [Google Scholar] [CrossRef]
  91. Voelz, K.; Lammas, D.A.; May, R.C. Cytokine Signaling Regulates the Outcome of Intracellular Macrophage Parasitism by Cryptococcus neoformans. Infect. Immun. 2009, 77, 3450–3457. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, Y.; Wang, F.; Tompkins, K.C.; McNamara, A.; Jain, A.V.; Moore, B.B.; Toews, G.B.; Huffnagle, G.B.; Olszewski, M.A. Robust Th1 and Th17 Immunity Supports Pulmonary Clearance but Cannot Prevent Systemic Dissemination of Highly Virulent Cryptococcus neoformans H99. Am. J. Pathol. 2009, 175, 2489–2500. [Google Scholar] [CrossRef]
  93. Hardison, S.E.; Ravi, S.; Wozniak, K.L.; Young, M.L.; Olszewski, M.A.; Wormley, F.L., Jr. Pulmonary Infection with an Interferon-γ-Producing Cryptococcus neoformans Strain Results in Classical Macrophage Activation and Protection. Am. J. Pathol. 2010, 176, 774–785. [Google Scholar] [CrossRef] [PubMed]
  94. Ruytinx, P.; Proost, P.; Van Damme, J.; Struyf, S. Chemokine-Induced Macrophage Polarization in Inflammatory Conditions. Front. Immunol. 2018, 9, 1930. [Google Scholar] [CrossRef] [PubMed]
  95. Hardison, S.E.; Herrera, G.; Young, M.L.; Hole, C.R.; Wozniak, K.L.; Wormley, F.L., Jr. Protective Immunity against Pulmonary Cryptococcosis Is Associated with STAT1-Mediated Classical Macrophage Activation. J. Immunol. 2012, 189, 4060–4068. [Google Scholar] [CrossRef]
  96. 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]
  97. 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]
  98. Leopold Wager, C.M.; Hole, C.R.; Campuzano, A.; Castro-Lopez, N.; Cai, H.; Caballero Van Dyke, M.C.; Wozniak, K.L.; Wang, Y.; Wormley, F.L., Jr. IFN-γ immune priming of macrophages in vivo induces prolonged STAT1 binding and protection against Cryptococcus neoformans. PLoS Pathog. 2018, 14, e1007358. [Google Scholar] [CrossRef]
  99. 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]
  100. Xiao, G.; Miyazato, A.; Inden, K.; Nakamura, K.; Shiratori, K.; Nakagawa, K.; Miyazawa, T.; Suzuki, K.; Kaku, M.; Kawakami, K. Cryptococcus neoformans inhibits nitric oxide synthesis caused by CpG-oligodeoxynucleotide-stimulated macrophages in a fashion independent of capsular polysaccharides. Microbiol. Immunol. 2008, 52, 171–179. [Google Scholar] [CrossRef]
  101. Naslund, P.K.; Miller, W.C.; Granger, D.L. Cryptococcus neoformans fails to induce nitric oxide synthase in primed murine macrophage-like cells. Infect. Immun. 1995, 63, 1298–1304. [Google Scholar] [CrossRef]
  102. Alspaugh, J.A.; Granger, D.L. Inhibition of Cryptococcus neoformans replication by nitrogen oxides supports the role of these molecules as effectors of macrophage-mediated cytostasis. Infect. Immun. 1991, 59, 2291–2296. [Google Scholar] [CrossRef] [PubMed]
  103. Aguirre, K.M.; Gibson, G.W. Differing requirement for inducible nitric oxide synthase activity in clearance of primary and secondary Cryptococcus neoformans infection. Med. Mycol. 2000, 38, 343–353. [Google Scholar] [CrossRef] [PubMed]
  104. Rivera, J.; Mukherjee, J.; Weiss, L.M.; Casadevall, A. Antibody Efficacy in Murine Pulmonary Cryptococcus neoformans Infection: A Role for Nitric Oxide. J. Immunol. 2002, 168, 3419–3427. [Google Scholar] [CrossRef] [PubMed]
  105. Stenzel, W.; Müller, U.; Köhler, 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]
  106. Hardison, S.E.; Wozniak, K.L.; Kolls, J.K.; Wormley, F.L., Jr. Interleukin-17 Is Not Required for Classical Macrophage Activation in a Pulmonary Mouse Model of Cryptococcus neoformans Infection. Infect. Immun. 2010, 78, 5341–5351. [Google Scholar] [CrossRef]
  107. Subramani, A.; Griggs, P.; Frantzen, N.; Mendez, J.; Tucker, J.; Murriel, J.; Sircy, L.M.; Millican, G.E.; McClelland, E.E.; Seipelt-Thiemann, R.L.; et al. Intracellular Cryptococcus neoformans disrupts the transcriptome profile of M1- and M2-polarized host macrophages. PLoS ONE 2020, 15, e0233818. [Google Scholar] [CrossRef]
  108. Nelson, B.N.; Daugherty, C.S.; Sharp, R.R.; Booth, J.L.; Patel, V.I.; Metcalf, J.P.; Jones, K.L.; Wozniak, K.L. Protective interaction of human phagocytic APC subsets with Cryptococcus neoformans induces genes associated with metabolism and antigen presentation. Front. Immunol. 2022, 13, 1054477. [Google Scholar] [CrossRef]
  109. Hansakon, A.; Mutthakalin, P.; Ngamskulrungroj, P.; Chayakulkeeree, M.; Angkasekwinai, P. Cryptococcus neoformans and Cryptococcus gattii clinical isolates from Thailand display diverse phenotypic interactions with macrophages. Virulence 2019, 10, 26–36. [Google Scholar] [CrossRef]
  110. 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]
  111. Tucker, S.C.; Casadevall, A. Replication of Cryptococcus neoformans in macrophages is accompanied by phagosomal permeabilization and accumulation of vesicles containing polysaccharide in the cytoplasm. Proc. Natl. Acad. Sci. USA 2002, 99, 3165–3170. [Google Scholar] [CrossRef] [PubMed]
  112. Johnston, S.A.; May, R.C. The Human Fungal Pathogen Cryptococcus neoformans Escapes Macrophages by a Phagosome Emptying Mechanism That Is Inhibited by Arp2/3 Complex-Mediated Actin Polymerisation. PLoS Pathog. 2010, 6, e1001041. [Google Scholar] [CrossRef] [PubMed]
  113. De Leon-Rodriguez, C.M.; Rossi, D.C.P.; Fu, M.S.; Dragotakes, Q.; Coelho, C.; Guerrero Ros, I.; Caballero, B.; Nolan, S.J.; Casadevall, A. The Outcome of the Cryptococcus neoformans—Macrophage Interaction Depends on Phagolysosomal Membrane Integrity. J. Immunol. 2018, 201, 583–603. [Google Scholar] [CrossRef]
  114. Boya, P.; Kroemer, G. Lysosomal membrane permeabilization in cell death. Oncogene 2008, 27, 6434–6451. [Google Scholar] [CrossRef]
  115. Davis, J.M.; Huang, M.; Botts, M.R.; Hull, C.M.; Huttenlocher, A. A Zebrafish Model of Cryptococcal Infection Reveals Roles for Macrophages, Endothelial Cells, and Neutrophils in the Establishment and Control of Sustained Fungemia. Infect. Immun. 2016, 84, 3047–3062. [Google Scholar] [CrossRef] [PubMed]
  116. Diamond, R.D.; Bennett, J.E. Growth of Cryptococcus neoformans Within Human Macrophages In Vitro. Infect. Immun. 1973, 7, 231–236. [Google Scholar] [CrossRef]
  117. Levitz, S.M.; Nong, S.-H.; Seetoo, K.F.; Harrison, T.S.; Speizer, R.A.; Simons, E.R. Cryptococcus neoformans Resides in an Acidic Phagolysosome of Human Macrophages. Infect. Immun. 1999, 67, 885–890. [Google Scholar] [CrossRef]
  118. Smith, L.M.; Dixon, E.F.; May, R.C. The fungal pathogen Cryptococcus neoformans manipulates macrophage phagosome maturation. Cell. Microbiol. 2014, 17, 702–713. [Google Scholar] [CrossRef] [PubMed]
  119. Noverr, M.C.; Cox, G.M.; Perfect, J.R.; Huffnagle, G.B. Role of PLB1 in Pulmonary Inflammation and Cryptococcal Eicosanoid Production. Infect. Immun. 2003, 71, 1538–1547. [Google Scholar] [CrossRef]
  120. Evans, R.J.; Li, Z.; Hughes, W.S.; Djordjevic, J.T.; Nielsen, K.; May, R.C. Cryptococcal Phospholipase B1 Is Required for Intracellular Proliferation and Control of Titan Cell Morphology during Macrophage Infection. Infect. Immun. 2015, 83, 1296–1304. [Google Scholar] [CrossRef]
  121. Liu, T.-B.; Xue, C. Fbp1-Mediated Ubiquitin-Proteasome Pathway Controls Cryptococcus neoformans Virulence by Regulating Fungal Intracellular Growth in Macrophages. Infect. Immun. 2014, 82, 557–568. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. Nicola, A.M.; Robertson, E.J.; Albuquerque, P.; Derengowski, L.D.S.; Casadevall, A. Nonlytic Exocytosis of Cryptococcus neoformans from Macrophages Occurs In Vivo and Is Influenced by Phagosomal pH. mBio 2011, 2, e00167-11. [Google Scholar] [CrossRef] [PubMed]
  124. Chayakulkeeree, M.; Johnston, S.A.; Oei, J.B.; Lev, S.; Williamson, P.R.; Wilson, C.F.; Zuo, X.; Leal, A.L.; Vainstein, M.H.; Meyer, W.; et al. SEC14 is a specific requirement for secretion of phospholipase B1 and pathogenicity of Cryptococcus neoformans. Mol. Microbiol. 2011, 80, 1088–1101. [Google Scholar] [CrossRef]
  125. Santiago-Tirado, F.H.; Doering, T.L. False friends: Phagocytes as Trojan horses in microbial brain infections. PLoS Pathog. 2017, 13, e1006680. [Google Scholar] [CrossRef]
  126. Santiago-Tirado, F.H.; Onken, M.D.; Cooper, J.A.; Klein, R.S.; Doering, T.L. Trojan Horse Transit Contributes to Blood-Brain Barrier Crossing of a Eukaryotic Pathogen. mBio 2017, 8, e02183-16. [Google Scholar] [CrossRef]
  127. Gottfredsson, M.; Perfect, J.R. Fungal Meningitis. Semin. Neurol. 2000, 20, 307–322. [Google Scholar] [CrossRef]
  128. Chang, Y.C.; Stins, M.F.; McCaffery, M.J.; Miller, G.F.; Pare, D.R.; Dam, T.; Paul-Satyaseela, M.; Kim, K.S.; Kwon-Chung, K.J. Cryptococcal Yeast Cells Invade the Central Nervous System via Transcellular Penetration of the Blood-Brain Barrier. Infect. Immun. 2004, 72, 4985–4995. [Google Scholar] [CrossRef]
  129. Liu, T.-B.; Perlin, D.S.; Xue, C. Molecular mechanisms of cryptococcal meningitis. Virulence 2012, 3, 173–181. [Google Scholar] [CrossRef]
  130. Hawkins, T. Understanding and managing the adverse effects of antiretroviral therapy. Antivir. Res. 2010, 85, 201–209. [Google Scholar] [CrossRef]
  131. Montessori, V.; Press, N.; Harris, M.; Akagi, L.; Montaner, J.S.G. Adverse effects of antiretroviral therapy for HIV infection. Can. Med. Assoc. J. 2004, 170, 229–238. [Google Scholar]
  132. Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting Microglial Cells Are Highly Dynamic Surveillants of Brain Parenchyma in Vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef]
  133. Kreutzberg, G.W. Microglia: A sensor for pathological events in the CNS. Trends Neurosci. 1996, 19, 312–318. [Google Scholar] [CrossRef]
  134. Kettenmann, H.; Hanisch, U.-K.; Noda, M.; Verkhratsky, A. Physiology of Microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef]
  135. Nau, R.; Ribes, S.; Djukic, M.; Eiffert, H. Strategies to increase the activity of microglia as efficient protectors of the brain against infections. Front. Cell. Neurosci. 2014, 8, 138. [Google Scholar] [CrossRef] [PubMed]
  136. Chhatbar, C.; Prinz, M. The roles of microglia in viral encephalitis: From sensome to therapeutic targeting. Cell. Mol. Immunol. 2021, 18, 250–258. [Google Scholar] [CrossRef] [PubMed]
  137. Rock, R.B.; Gekker, G.; Hu, S.; Sheng, W.S.; Cheeran, M.; Lokensgard, J.R.; Peterson, P.K. Role of Microglia in Central Nervous System Infections. Clin. Microbiol. Rev. 2004, 17, 942–964. [Google Scholar] [CrossRef]
  138. Waltl, I.; Kalinke, U. Beneficial and detrimental functions of microglia during viral encephalitis. Trends Neurosci. 2022, 45, 158–170. [Google Scholar] [CrossRef] [PubMed]
  139. Buchanan, K.L.; Doyle, H.A. Requirement for CD4+ T Lymphocytes in Host Resistance against Cryptococcus neoformans in the Central Nervous System of Immunized Mice. Infect. Immun. 2000, 68, 456–462. [Google Scholar] [CrossRef]
  140. Chao, C.C.; Hu, S.; Molitor, T.W.; Shaskan, E.G.; Peterson, P.K. Activated microglia mediate neuronal cell injury via a nitric oxide mechanism. J. Immunol. 1992, 149, 2736–2741. [Google Scholar] [CrossRef]
  141. Blasi, E.; Barluzzi, R.; Mazzolla, R.; Tancini, B.; Saleppico, S.; Puliti, M.; Pitzurra, L.; Bistoni, F. Role of nitric oxide and melanogenesis in the accomplishment of anticryptococcal activity by the BV-2 microglial cell line. J. Neuroimmunol. 1995, 58, 111–116. [Google Scholar] [CrossRef] [PubMed]
  142. Adami, C.; Sorci, G.; Blasi, E.; Agneletti, A.L.; Bistoni, F.; Donato, R. S100B expression in and effects on microglia. Glia 2001, 33, 131–142. [Google Scholar] [CrossRef] [PubMed]
  143. Sun, D.; Sun, P.; Li, H.; Zhang, M.; Liu, G.; Strickland, A.B.; Chen, Y.; Fu, Y.; Xu, J.; Yosri, M.; et al. Fungal dissemination is limited by liver macrophage filtration of the blood. Nat. Commun. 2019, 10, 4566. [Google Scholar] [CrossRef] [PubMed]
  144. Heung, L.J.; Hohl, T.M. Inflammatory monocytes are detrimental to the host immune response during acute infection with Cryptococcus neoformans. PLoS Pathog. 2019, 15, e1007627. [Google Scholar] [CrossRef]
  145. Espinosa, V.; Jhingran, A.; Dutta, O.; Kasahara, S.; Donnelly, R.; Du, P.; Rosenfeld, J.; Leiner, I.; Chen, C.-C.; Ron, Y.; et al. Inflammatory Monocytes Orchestrate Innate Antifungal Immunity in the Lung. PLoS Pathog. 2014, 10, e1003940. [Google Scholar] [CrossRef]
  146. Hohl, T.M.; Rivera, A.; Lipuma, L.; Gallegos, A.; Shi, C.; Mack, M.; Pamer, E.G. Inflammatory Monocytes Facilitate Adaptive CD4 T Cell Responses during Respiratory Fungal Infection. Cell Host Microbe 2009, 6, 470–481. [Google Scholar] [CrossRef] [PubMed]
  147. Wüthrich, M.; Ersland, K.; Sullivan, T.; Galles, K.; Klein, B.S. Fungi Subvert Vaccine T Cell Priming at the Respiratory Mucosa by Preventing Chemokine-Induced Influx of Inflammatory Monocytes. Immunity 2012, 36, 680–692. [Google Scholar] [CrossRef]
  148. Ngo, L.Y.; Kasahara, S.; Kumasaka, D.K.; Knoblaugh, S.E.; Jhingran, A.; Hohl, T.M. Inflammatory Monocytes Mediate Early and Organ-Specific Innate Defense During Systemic Candidiasis. J. Infect. Dis. 2014, 209, 109–119. [Google Scholar] [CrossRef]
  149. Szymczak, W.A.; Deepe, G.S., Jr. The CCL7-CCL2-CCR2 Axis Regulates IL-4 Production in Lungs and Fungal Immunity. J. Immunol. 2009, 183, 1964–1974. [Google Scholar] [CrossRef]
  150. 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]
  151. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef]
  152. Gomez Perdiguero, E.; Klapproth, K.; Schulz, C.; Busch, K.; Azzoni, E.; Crozet, L.; Garner, H.; Trouillet, C.; de Bruijn, M.F.; Geissmann, F.; et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 2015, 518, 547–551. [Google Scholar] [CrossRef] [PubMed]
  153. Van De Laar, L.; Saelens, W.; De Prijck, S.; Martens, L.; Scott, C.L.; Van Isterdael, G.; Hoffmann, E.; Beyaert, R.; Saeys, Y.; Lambrecht, B.N.; et al. Yolk Sac Macrophages, Fetal Liver, and Adult Monocytes Can Colonize an Empty Niche and Develop into Functional Tissue-Resident Macrophages. Immunity 2016, 44, 755–768. [Google Scholar] [CrossRef] [PubMed]
  154. Davies, L.C.; Jenkins, S.J.; Allen, J.E.; Taylor, P.R. Tissue-resident macrophages. Nat. Immunol. 2013, 14, 986–995. [Google Scholar] [CrossRef]
  155. Hou, F.; Xiao, K.; Tang, L.; Xie, L. Diversity of Macrophages in Lung Homeostasis and Diseases. Front. Immunol. 2021, 12, 753940. [Google Scholar] [CrossRef] [PubMed]
  156. Morales-Nebreda, L.; Misharin, A.V.; Perlman, H.; Budinger, G.R. The heterogeneity of lung macrophages in the susceptibility to disease. Eur. Respir. Rev. 2015, 24, 505–509. [Google Scholar] [CrossRef]
  157. Guilliams, M.; De Kleer, I.; Henri, S.; Post, S.; Vanhoutte, L.; De Prijck, S.; Deswarte, K.; Malissen, B.; Hammad, H.; Lambrecht, B.N. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 2013, 210, 1977–1992. [Google Scholar] [CrossRef]
  158. Yona, S.; Kim, K.-W.; Wolf, Y.; Mildner, A.; Varol, D.; Breker, M.; Strauss-Ayali, D.; Viukov, S.; Guilliams, M.; Misharin, A.; et al. Fate Mapping Reveals Origins and Dynamics of Monocytes and Tissue Macrophages under Homeostasis. Immunity 2013, 38, 79–91. [Google Scholar] [CrossRef]
  159. Gibbings, S.L.; Thomas, S.M.; Atif, S.M.; McCubbrey, A.L.; Desch, A.N.; Danhorn, T.; Leach, S.M.; Bratton, D.L.; Henson, P.M.; Janssen, W.J.; et al. Three Unique Interstitial Macrophages in the Murine Lung at Steady State. Am. J. Respir. Cell Mol. Biol. 2017, 57, 66–76. [Google Scholar] [CrossRef]
  160. Gautier, E.L.; Shay, T.; Miller, J.; Greter, M.; Jakubzick, C.; Ivanov, S.; Helft, J.; Chow, A.; Elpek, K.G.; Gordonov, S.; et al. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 2012, 13, 1118–1128. [Google Scholar] [CrossRef]
  161. Tan, S.Y.S.; Krasnow, M.A. Developmental origin of lung macrophage diversity. Development 2016, 143, 1318–1327. [Google Scholar] [CrossRef] [PubMed]
  162. Hey, Y.-Y.; Tan, J.K.; O’Neill, H.C. Redefining Myeloid Cell Subsets in Murine Spleen. Front. Immunol. 2015, 6, 652. [Google Scholar] [CrossRef] [PubMed]
  163. Lam, J.S.; Huang, H.; Levitz, S.M. Effect of Differential N-linked and O-linked Mannosylation on Recognition of Fungal Antigens by Dendritic Cells. PLoS ONE 2007, 2, e1009. [Google Scholar] [CrossRef]
  164. Lam, J.S.; Mansour, M.K.; Specht, C.A.; Levitz, S.M. A Model Vaccine Exploiting Fungal Mannosylation to Increase Antigen Immunogenicity. J. Immunol. 2005, 175, 7496–7503. [Google Scholar] [CrossRef]
  165. Hewitt, E.W. The MHC class I antigen presentation pathway: Strategies for viral immune evasion. Immunology 2003, 110, 163–169. [Google Scholar] [CrossRef] [PubMed]
  166. Silva, M.T. Neutrophils and macrophages work in concert as inducers and effectors of adaptive immunity against extracellular and intracellular microbial pathogens. J. Leukoc. Biol. 2010, 87, 805–813. [Google Scholar] [CrossRef] [PubMed]
  167. Campuzano, A.; Wormley, F.L., Jr. Innate Immunity against Cryptococcus, from Recognition to Elimination. J. Fungi 2018, 4, 33. [Google Scholar] [CrossRef]
  168. Chiller, T.; Farrokhshad, K.; Brummer, E.; Stevens, D.A. Effect of granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor on polymorphonuclear neutrophils, monocytes or monocyte-derived macrophages combined with voriconazole against Cryptococcus neoformans. Med. Mycol. 2002, 40, 21–26. [Google Scholar] [CrossRef]
  169. Mayadas, T.N.; Cullere, X.; Lowell, C.A. The Multifaceted Functions of Neutrophils. Annu. Rev. Pathol. Mech. Dis. 2014, 9, 181–218. [Google Scholar] [CrossRef]
  170. 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]
  171. Miller, M.F.; Mitchell, T.G. Killing of Cryptococcus neoformans strains by human neutrophils and monocytes. Infect. Immun. 1991, 59, 24–28. [Google Scholar] [CrossRef]
  172. Lämmermann, T. In the eye of the neutrophil swarm—Navigation signals that bring neutrophils together in inflamed and infected tissues. J. Leukoc. Biol. 2015, 100, 55–63. [Google Scholar] [CrossRef]
  173. 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. 2016, 84, 216–229. [Google Scholar] [CrossRef] [PubMed]
  174. Voelz, K.; May, R.C. Cryptococcal Interactions with the Host Immune System. Eukaryot. Cell 2010, 9, 835–846. [Google Scholar] [CrossRef]
  175. 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]
  176. 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]
  177. Roubey, R.A.; Ross, G.D.; Merrill, J.T.; Walton, F.; Reed, W.; Winchester, R.J.; Buyon, J.P. Staurosporine inhibits neutrophil phagocytosis but not iC3b binding mediated by CR3 (CD11b/CD18). J Immunol. 1991, 146, 3557–3562. [Google Scholar] [CrossRef]
  178. Podolnikova, N.P.; Kushchayeva, Y.S.; Wu, Y.; Faust, J.; Ugarova, T.P. The Role of Integrins αMβ2 (Mac-1, CD11b/CD18) and αDβ2 (CD11d/CD18) in Macrophage Fusion. Am. J. Pathol. 2016, 186, 2105–2116. [Google Scholar] [CrossRef]
  179. Filippi, M.-D. Neutrophil transendothelial migration: Updates and new perspectives. Blood 2019, 133, 2149–2158. [Google Scholar] [CrossRef] [PubMed]
  180. Kumar, S.; Xu, J.; Perkins, C.; Guo, F.; Snapper, S.; Finkelman, F.D.; Zheng, Y.; Filippi, M.-D. Cdc42 regulates neutrophil migration via crosstalk between WASp, CD11b, and microtubules. Blood 2012, 120, 3563–3574. [Google Scholar] [CrossRef] [PubMed]
  181. Cara, D.C.; Kaur, J.; Forster, M.; McCafferty, D.-M.; Kubes, P. Role of p38 Mitogen-Activated Protein Kinase in Chemokine-Induced Emigration and Chemotaxis In Vivo. J. Immunol. 2001, 167, 6552–6558. [Google Scholar] [CrossRef]
  182. Kim, D.; Haynes, C.L. The role of p38 MAPK in neutrophil functions: Single cell chemotaxis and surface marker expression. Analyst 2013, 138, 6826–6833. [Google Scholar] [CrossRef]
  183. Liu, X.; Ma, B.; Malik, A.B.; Tang, H.; Yang, T.; Sun, B.; Wang, G.; Minshall, R.D.; Li, Y.; Zhao, Y.; et al. Bidirectional regulation of neutrophil migration by mitogen-activated protein kinases. Nat. Immunol. 2012, 13, 457–464. [Google Scholar] [CrossRef] [PubMed]
  184. Chen, M.; Lam, B.K.; Kanaoka, Y.; Nigrovic, P.A.; Audoly, L.P.; Austen, K.F.; Lee, D.M. Neutrophil-derived leukotriene B4 is required for inflammatory arthritis. J. Exp. Med. 2006, 203, 837–842. [Google Scholar] [CrossRef]
  185. Majumdar, R.; Tavakoli Tameh, A.; Arya, S.B.; Parent, C.A. Exosomes mediate LTB4 release during neutrophil chemotaxis. PLoS Biol. 2021, 19, e3001271. [Google Scholar] [CrossRef]
  186. Ng, L.G.; Qin, J.S.; Roediger, B.; Wang, Y.; Jain, R.; Cavanagh, L.L.; Smith, A.L.; Jones, C.A.; de Veer, M.; Grimbaldeston, M.A.; et al. Visualizing the Neutrophil Response to Sterile Tissue Injury in Mouse Dermis Reveals a Three-Phase Cascade of Events. J. Investig. Dermatol. 2011, 131, 2058–2068. [Google Scholar] [CrossRef]
  187. Rocha, J.D.B.; Nascimento, M.T.C.; Decote-Ricardo, D.; Côrte-Real, S.; Morrot, A.; Heise, N.; Nunes, M.P.; Previato, J.O.; Mendonça-Previato, L.; DosReis, G.A.; et al. Capsular polysaccharides from Cryptococcus neoformans modulate production of neutrophil extracellular traps (NETs) by human neutrophils. Sci. Rep. 2015, 5, 8008. [Google Scholar] [CrossRef] [PubMed]
  188. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
  189. Ellerbroek, P.M.; Walenkamp, A.M.; Hoepelman, A.I.; Coenjaerts, F.E. Effects of the capsular polysaccharides of Cryptococcus neoformans on phagocyte migration and inflammatory mediators. Curr. Med. Chem. 2004, 11, 253–266. [Google Scholar] [CrossRef]
  190. Michaud, J.-P.; Bellavance, M.-A.; Préfontaine, P.; Rivest, S. Real-Time In Vivo Imaging Reveals the Ability of Monocytes to Clear Vascular Amyloid Beta. Cell Rep. 2013, 5, 646–653. [Google Scholar] [CrossRef] [PubMed]
  191. Bryan, A.M.; Del Poeta, M.; Luberto, C. Sphingolipids as Regulators of the Phagocytic Response to Fungal Infections. Mediat. Inflamm. 2015, 2015, 640540. [Google Scholar] [CrossRef]
  192. 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]
  193. Musubire, A.K.; Meya, D.B.; Rhein, J.; Meintjes, G.; Bohjanen, P.R.; Nuwagira, E.; Muzoora, C.; Boulware, D.R.; Hullsiek, K.H.; COAT and ASTRO Trial Teams. Blood neutrophil counts in HIV-infected patients with cryptococcal meningitis: Association with mortality. PLoS ONE 2018, 13, e0209337. [Google Scholar] [CrossRef] [PubMed]
  194. Zhang, M.; Sun, D.; Shi, M. Dancing cheek to cheek: Cryptococcus neoformans and phagocytes. Springerplus 2015, 4, 410. [Google Scholar] [CrossRef] [PubMed]
  195. Chen, Y.; Shi, Z.W.; Strickland, A.B.; Shi, M. Cryptococcus neoformans Infection in the Central Nervous System: The Battle between Host and Pathogen. J. Fungi 2022, 8, 1069. [Google Scholar] [CrossRef] [PubMed]
  196. Watkins, R.A.; Andrews, A.; Wynn, C.; Barisch, C.; King, J.S.; Johnston, S.A. Cryptococcus neoformans Escape From Dictyostelium Amoeba by Both WASH-Mediated Constitutive Exocytosis and Vomocytosis. Front. Cell. Infect. Microbiol. 2018, 8, 108. [Google Scholar] [CrossRef]
  197. Chrisman, C.J.; Alvarez, M.; Casadevall, A. Phagocytosis of Cryptococcus neoformans by, and Nonlytic Exocytosis from, Acanthamoeba castellanii. Appl. Environ. Microbiol. 2010, 76, 6056–6062. [Google Scholar] [CrossRef]
Figure 1. PRRs and CLRs Involved in Cryptococcal Recognition by DCs. DCs are involved in the recognition, engulfment, and killing of pathogens. Initial recognition involves the use of either intra- or extracellular pattern recognition receptors (PPRs), as well as C-type lectin receptors (CLRs). After recognition of their corresponding PAMPS, a cascade of signaling is triggered to initiate the maturation of the DCs and CD-mediated T cell activation. (A) While not all TLRs are required for the development of a protective immune response against C. neoformans, the intracellular TLR9 is required. TLR9 recognizes URA5 C. neoformans DNA after degradation of an engulfed cryptococcal cell. It then initiates a signal cascade involving the adaptor molecule MyD88, which is also a requirement for the initiation of DC maturation and a protective response. (B) While not as well studied, some CLRs have been shown to be necessary for the recognition and phagocytosis of C. neoformans by DCs. Without the presence of Dectin-2, the signaling molecule SyK, or the adaptor molecule CARD9, there is a reduction in the phagocytosis of cryptococcal cells by DCs. Within pDCs, the requirement of Dectin-3 is dependent on the host.
Figure 1. PRRs and CLRs Involved in Cryptococcal Recognition by DCs. DCs are involved in the recognition, engulfment, and killing of pathogens. Initial recognition involves the use of either intra- or extracellular pattern recognition receptors (PPRs), as well as C-type lectin receptors (CLRs). After recognition of their corresponding PAMPS, a cascade of signaling is triggered to initiate the maturation of the DCs and CD-mediated T cell activation. (A) While not all TLRs are required for the development of a protective immune response against C. neoformans, the intracellular TLR9 is required. TLR9 recognizes URA5 C. neoformans DNA after degradation of an engulfed cryptococcal cell. It then initiates a signal cascade involving the adaptor molecule MyD88, which is also a requirement for the initiation of DC maturation and a protective response. (B) While not as well studied, some CLRs have been shown to be necessary for the recognition and phagocytosis of C. neoformans by DCs. Without the presence of Dectin-2, the signaling molecule SyK, or the adaptor molecule CARD9, there is a reduction in the phagocytosis of cryptococcal cells by DCs. Within pDCs, the requirement of Dectin-3 is dependent on the host.
Jof 09 00617 g001
Figure 2. Two Distinct Outcomes of Interactions with Human Innate Phagocytes. Following recognition of cryptococcal cells within the alveoli of the lungs by the host’s pulmonary phagocytes, the innate immune cells are involved in the removal of the pathogen and clearance of the cryptococcal infection. There are two distinct outcomes of the cryptococcal–host interaction. Once C. neoformans cells have been internalized, some cells are capable of actively killing the fungal cells, resulting in clearance of the cryptococcal infection from the host. However, a few cell types/subsets have been shown to be permissive to cryptococcal growth, allowing for intracellular growth and ultimately the trafficking of cryptococcal cells to the central nervous system. These cells are able to transport the fungal cells across the blood–brain barrier (BBB), resulting in the development of cryptococcal meningitis.
Figure 2. Two Distinct Outcomes of Interactions with Human Innate Phagocytes. Following recognition of cryptococcal cells within the alveoli of the lungs by the host’s pulmonary phagocytes, the innate immune cells are involved in the removal of the pathogen and clearance of the cryptococcal infection. There are two distinct outcomes of the cryptococcal–host interaction. Once C. neoformans cells have been internalized, some cells are capable of actively killing the fungal cells, resulting in clearance of the cryptococcal infection from the host. However, a few cell types/subsets have been shown to be permissive to cryptococcal growth, allowing for intracellular growth and ultimately the trafficking of cryptococcal cells to the central nervous system. These cells are able to transport the fungal cells across the blood–brain barrier (BBB), resulting in the development of cryptococcal meningitis.
Jof 09 00617 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Conn, B.N.; Wozniak, K.L. Innate Pulmonary Phagocytes and Their Interactions with Pathogenic Cryptococcus Species. J. Fungi 2023, 9, 617. https://doi.org/10.3390/jof9060617

AMA Style

Conn BN, Wozniak KL. Innate Pulmonary Phagocytes and Their Interactions with Pathogenic Cryptococcus Species. Journal of Fungi. 2023; 9(6):617. https://doi.org/10.3390/jof9060617

Chicago/Turabian Style

Conn, Brittney N., and Karen L. Wozniak. 2023. "Innate Pulmonary Phagocytes and Their Interactions with Pathogenic Cryptococcus Species" Journal of Fungi 9, no. 6: 617. https://doi.org/10.3390/jof9060617

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