- Research
- Open access
- Published:
IL-6 deficiency accelerates cerebral cryptococcosis and alters glial cell responses
Journal of Neuroinflammation volumeĀ 21, ArticleĀ number:Ā 242 (2024)
Abstract
Cryptococcus neoformans (Cn) is an opportunistic encapsulated fungal pathogen that causes life-threatening meningoencephalitis in immunosuppressed individuals. Since IL-6 is important for blood-brain barrier support and its deficiency has been shown to facilitate Cn brain invasion, we investigated the impact of IL-6 on systemic Cn infection in vivo, focusing on central nervous system (CNS) colonization and glial responses, specifically microglia and astrocytes. IL-6 knock-out (IL-6ā/ā) mice showed faster mortality than C57BL/6 (Wild-type) and IL-6ā/ā supplemented with recombinant IL-6 (rIL-6; 40 pg/g/day) mice. Despite showing early lung inflammation but no major histological differences in pulmonary cryptococcosis progression among the experimental groups, IL-6ā/ā mice had significantly higher blood and brain tissue fungal burden at 7-days post infection. Exposure of cryptococci to rIL-6 in vitro increased capsule growth. In addition, IL-6ā/ā brains were characterized by an increased dystrophic microglia number during Cn infection, which are associated with neurodegeneration and senescence. In contrast, the brains of IL-6-producing or -supplemented mice displayed high numbers of activated and phagocytic microglia, which are related to a stronger anti-cryptococcal response or tissue repair. Likewise, culture of rIL-6 with microglia-like cells promoted high fungal phagocytosis and killing, whereas IL-6 silencing in microglia decreased fungal phagocytosis. Lastly, astrogliosis was high and moderate in infected brains removed from Wild-type and IL-6ā/ā supplemented with rIL-6 animals, respectively, while minimal astrogliosis was observed in IL-6ā/ā tissue, highlighting the potential of astrocytes in containing and combating cryptococcal infection. Our findings suggest a critical role for IL-6 in Cn CNS dissemination, neurocryptococcosis development, and host defense.
Introduction
Cryptococcus neoformans (Cn) is an encapsulated yeast-like fungus that causes opportunistic infection mostly in immunocompromised patients and occasionally in healthy individuals. Most cases are reported in sub-Saharan Africa where HIV is highly prevalent and access to optimal anti-cryptococcal therapy is inadequate. Cn establishes infection in the lungs upon inhalation of fungal spores or desiccated yeasts [1], enters the bloodstream during immunosuppression, and invades the central nervous system (CNS) transcellularly [2], paracellularly [3], or using phagocytes as a Trojan horse [4] typically resulting in life-threatening meningoencephalitis. Cerebral cryptococcosis is the third most common CNS infection in patients with HIV/AIDS [5]. The mortality rate of Cn infection is high (~ā74%), accounting for 112,000 deaths annually worldwide [6]. The polysaccharide capsule is a major contributor to Cn virulence [7]. Glucuronoxylomannan (GXM), the main capsular component, is extensively secreted and accumulates considerably in serum and cerebrospinal fluid (CSF) during infection [7, 8], contributing significantly to Cn pathogenesis [9]. For instance, GXM stimulates HIV proliferation [10] and weakens the host immunity by interfering with phagocytosis, antigen presentation, leukocyte migration, and specific antibody (Ab) responses [9].
Anti-cryptococcal response in the CNS is weak or delayed compared to that in peripheral organs, suggesting that effective immunity may involve the activation of T cells and their eventual entry into the CNS [11]. Astrocytic proliferation and microglial activation in association with neuronal damage and reduced cellular repair are characteristics of cryptococcal meningoencephalitis [12]. Post-mortem neuropathological examinations of patientsā brains have demonstrated that Cn GXM is ingested and localizes inside of microglia [13, 14]. Intracellular and extracellular defense against fungi by microglia depends on cytokine release, such as interferon (IFN)-Ī³, complement activation [15], and opsonization of antigens [16]. For instance, nitric oxide (NO) production is stimulated in microglia after its S100B protein surrounds the phagosome of opsonized Cn in the presence of IFN-Ī³ [17]. Cn GXM is also commonly associated with reactive astrocytes [11], which accumulate around cryptococcomas during infection [13]. We recently demonstrated a correlation between glia distribution and GXM localization that varies depending on brain region of infection in mice [12]. Hence, expanding the gap of knowledge on the involvement of glia cells on the immune responses against Cn CNS infection is imperative.
IL-6 has pleiotropic effects, including involvement in Ab production, B cell differentiation, T cell activation, and induction of acute phase proteins [18]. IL-6 stimulation of the hypothalamic-pituitary-adrenal axis serves to control the inflammatory response. Thus, IL-6 also has significant indirect anti-inflammatory properties [19]. Elevated levels of IL-6 may have a direct pathogenic role in neurodegenerative diseases such as Alzheimerās disease and multiple sclerosis [20]. High IL-6 levels are associated with HIV infection [21]. Lack of IL-6 increases blood-brain barrier (BBB) permeability after cryptococcal pulmonary infection [22]. IL-6 is reduced in plasma of individuals with AIDS, and this is related to fungemia and dissemination [23]. Intracerebral challenge of mice with exogenous IL-6 enhances survival during cryptococcal infection by reducing fungal load in blood and brain [24]. Toll-like receptor stimulation increases phagocytosis of Cn by microglial cells and cytokine production, including IL-6 [25]. Thus, the role of IL-6 in CNS invasion and colonization, and particularly its impact on glial cells has not been extensively explored.
In this study, we investigated the impact of IL-6 on systemic Cn infection in vivo, with emphasis on CNS colonization and glial responses, especially microglia and astrocytes. We compared the survivability and pathophysiology of disseminated cryptococcosis in Wild-type, IL-6ā/ā, and IL-6ā/ā mice supplemented with exogenous IL-6. We also provided significant evidence demonstrating that IL-6 is an important immunomodulator and influences glial responses and effector functions against cryptococcal infection. In contrast, Cn exposure to IL-6 induces capsule enlargement, which may have significant implications in the progression of cryptococcal meningoencephalitis. Our findings provide insight into the dynamic role of IL-6 in neurocryptococcosis and offer novel research avenues in the study of Cn pathogenesis.
Results
IL-6 deficiency reduces survival in mice systemically infected with Cn
To evaluate the importance of IL-6 in controlling cryptococcosis, we infected Wild-type C57BL/6, IL-6ā/ā, and IL-6ā/ā supplemented with recombinant (r) IL-6 (40 pg/g/day) mice with Cn strain H99 cells (Fig.Ā 1A). IL-6ā/ā rodents (median survival: 7-days post infection [dpi]) showed significantly faster mortality than Wild-type- and IL-6ā/ā + rIL-6-treated mice (median survival: 8- and 9-dpi, respectively; Pā<ā0.001; Fig.Ā 1B). In addition, IL-6ā/ā + rIL-6-treated mice showed prolonged survivability than Wild-type mice (Pā<ā0.001; Fig.Ā 1B), with the last animal dying at 14-dpi versus 10-dpi, respectively.
Since IL-6 is involved in temperature regulation in mammals, we monitored changes in core body temperature of Wild-type, IL-6ā/ā, and IL-6ā/ā + rIL-6 mice after Cn infection. Animals in all groups showed a 0.5ĀŗC decrease in their body temperature a day after the rIL-6 pre-infection dose (Fig.Ā 1C). IL-6ā/ā mice had a considerable temperature drop 6-dpi and correlated with their median mortality. Similarly, Wild-type mice showed a substantial core temperature reduction at 7-dpi. Interestingly, IL-6ā/ā + rIL-6 mice displayed an approximately 0.5ĀŗC body temperature increase at 2-dpi that was sustained until 5-dpi before fluctuating between 35ā36ĀŗC until most animals were dead.
Lastly, we also monitored weight loss in Wild-type, IL-6ā/ā, and IL-6ā/ā + rIL-6 mice after Cn infection as an indicator of disease progression and mortality (Fig.Ā 1D). All the groups showed a similar weight loss trend post-infection indicating that IL-6 deficiency had no impact on body weight loss during Cn infection.
Our findings indicate that IL-6 extends mice survival during Cn infection and is particularly important in maintaining an optimal temperature.
IL-6 modulates fungal proliferation during infection
We investigated the importance of IL-6 in systemic Cn disease using colony forming units (CFU) determinations and histopathological examinations of lung and brain tissue removed from Cn-infected-Wild-type, -IL-6ā/ā, and -IL-6ā/ā + rIL-6 mice at 3- and 7-dpi. At 3-dpi, IL-6ā/ā + rIL-6 mice (lungs, 3.33āĆā103 CFU/g; brain, 1.56āĆā105 CFU/g) had significantly higher cryptococcal load than Wild-type (lungs, 1.57āĆā103 CFU/g; Pā<ā0.0001; brain, 1.2āĆā105 CFU/g; Pā<ā0.001) and IL-6ā/ā (lungs, 2.1āĆā103 CFU/g; Pā<ā0.001; brain, 1.2āĆā105 CFU/g; Pā<ā0.001) mice, respectively (Fig.Ā 2A-C). The lungs and brains of Wild-type- and IL-6ā/ā-infected mice showed no difference in fungal burden. Notably, Wild-type mice showed minimal yeast cells in circulation relative to the IL-6ā/ā (1.92āĆā102 CFU/0.1 mL, Pā<ā0.01) and IL-6ā/ā + rIL-6 (2.5āĆā102 CFU/0.1 mL, Pā<ā0.01) mice that had similar blood load (Fig.Ā 2B).
Coronal lung tissue sections were stained with hematoxylin-eosin to examine host tissue morphological changes during cryptococcal infection (SFig.Ā 1). Representative pulmonary tissue removed from Wild-type mice (left panels) at 3-dpi displayed localized inflammation (4X) characterized by atelectasis (complete or partial collapse of a lung area) and alveolar emphysema (yellow arrow; 10X). In contrast, in regions of the lung with less inflammation, intrapulmonary bronchioles were also lined by normal epithelium (black arrow; 10X; SFig.Ā 1Ā A). A high magnification (20X) image exhibited pulmonary emphysematous changes and normal pseudostratified columnar ciliated epithelium lining the bronchioles. IL-6ā/ā lungs (middle panels) displayed more pronounced inflammation (4X) with considerable atelectasis (yellow arrow), diffuse thickening of interstitial tissue, and intrapulmonary bronchioles with severe hyperplasia of lining epithelium (blue arrow; 10X; SFig.Ā 1Ā A). High power magnification (20X) of the IL-6ā/ā-infected tissue demonstrated marked thickening of interstitial tissue with severe cell infiltration and severe hyperplasia of the epithelium lining of intrapulmonary bronchiole forming papillary like projections into the lumen (blue arrow). Pulmonary tissue from IL-6ā/ā + rIL-6 (right panels) evinced more and less inflammation (4X) than that observed in Wild-type and IL-6 deficient lungs, respectively. High magnification images (10-20X) exhibited alveoli with mild thickening of interstitial tissue (yellow arrow; SFig.Ā 1Ā A) and simple hyperplasia of the epithelium lining the intrapulmonary bronchioles (blue arrow). Remarkably, there was no visible cryptococci nor cryptococcoma formation in the lungs of the animals in any of the groups at this stage (3-dpi) of the infection.
Sagittal brain tissue sections (nā=ā3 mice/group/day) were stained with mucicarmine to identify the morphology of Cn in the host tissue during infection (Fig.Ā 2D). Representative 2X brain tissue sections from Wild-type, IL-6ā/ā, and IL-6ā/ā + rIL-6 mice show minimal differences in fungal colonization at 3-dpi (top panels). Higher magnification (4X; bottom panels) images of Wild-type, IL-6ā/ā, and IL-6ā/ā + rIL-6 brains displayed small brain cryptococcomas (black arrows).
On day 7 after infection, the lungs of IL-6ā/ā + rIL-6 mice (1.44āĆā106 CFU/g) had significantly higher fungal burden than Wild-type (4.09āĆā105 CFU/g; Pā<ā0.01) and IL-6ā/ā (3.92āĆā105 CFU/g; Pā<ā0.01) mice, respectively (Fig.Ā 2E). However, there were no differences in histopathology in the infected lungs of the compared groups. In fact, lung tissue removed from animals in each experimental group exhibited significant inflammation and cryptococcoma formation (green arrows; 4X; SFig.Ā 1B). They also showed alveoli with atelectasis and thickening of the interalveolar septa (yellow arrows; 10X). Higher magnification of the pulmonary tissue revealed bronchus-associated lymphoid tissue around cryptococcomas in all mice groups (red arrow heads; 20X). Moreover, IL-6ā/ā (blood, 1.7āĆā104 CFU/0.1 mL; brain, 3.96āĆā107 CFU/g) mice had a significantly higher fungal load than Wild-type (blood, 1.5āĆā103 CFU/0.1 mL, Pā<ā0.0001; brain, 2.8āĆā107 CFU/g, Pā<ā0.0001) and IL-6ā/ā + rIL-6 (blood, 1.4āĆā103 CFU/0.1 mL, Pā<ā0.0001; brain, 3.1āĆā107 CFU/g, Pā<ā0.001) mice in blood (Fig.Ā 2F) and brain (Fig.Ā 2G). There were no differences in fungal load in blood and brain in Wild-type and IL-6ā/ā + rIL-6 mice.
A representative 2X brain tissue section from a Wild-type mouse euthanized at 7-dpi showed multiple well-circumscribed areas of encephalomalacia or cryptococcomas (black arrows) filled and surrounded with GXM in the cerebral cortex, mid brain, and cerebellum (top left panel; Fig.Ā 2H). A 4X image (bottom left panel) shows the red staining (red arrow in the parenchyma) that extends from cryptococcoma (black arrow) to the cerebral cortex and diffuses to the leptomeninges and subarachnoid space (red arrows). A 2X section from an IL-6ā/ā mouse displays the mid brain with a well-defined cryptococcoma (black arrows; top middle panel). Additionally, the lumen and walls of the fourth ventricle evinced intense accumulation of mucicarmine staining (red arrow). A high magnification (4X) image demonstrates localized red staining indicative of cryptococci or GXM accumulation in the ependymal and sub-ependymal space lining of the fourth ventricle (red arrow; bottom middle panel). Finally, an IL-6ā/ā + rIL-6 brain section showed a few cryptococcomas (black arrows) with mucicarmine staining limited to their periphery (top right panel). Unlike the Wild-type and IL-6ā/ā brains, those supplemented with rIL-6 exhibited minimal mucicarmine staining or cryptococcal accumulation in the ependymal lining of the fourth ventricle (red arrows; bottom right panel).
Overall, our findings indicate that exogenous administration of IL-6 during early (3-dpi) systemic infection increases fungal burden in lungs, blood, and brain. However, as the infection progresses (7-dpi), IL-6ā/ā mice have higher fungal load in circulation and brain, suggesting the importance of this cytokine in Cn systemic dissemination and CNS colonization.
IL-6ā/ā mice showed increased Cn GXM accumulation in brain tissue
Cn GXM is copiously released during infection causing severe effects to the host immunity. Therefore, we evaluated how IL-6 deficiency impacted GXM accumulation in tissue during cerebral cryptococcosis (Fig.Ā 3). GXM was brown-stained in brain tissue using the specific monoclonal Ab (MAb) 18B7 (Fig.Ā 3A). The GXM intensity in different regions [cortex (Fig.Ā 3B), hippocampus (Fig.Ā 3C), hypothalamus (Fig.Ā 3D), midbrain (Fig.Ā 3E), and cerebellum (Fig.Ā 3F)] of sagittal brain tissue sections was blindly analyzed by three independent investigators (nā=ā10ā15 fields per brain region; nā=ā3 mice per group) using NIH ImageJ color deconvolution tool software. Immunolabeling of IL-6ā/ā-infected brain tissue demonstrated extensive GXM accumulation that diffused uniformly throughout the different brain regions (Fig.Ā 3A, middle panels). However, Wild-type mice evinced scattered GXM intensity with most of the polysaccharide accumulating in the mid brain, brainstem, and cerebellum and with lesser extent in the prefrontal cortex (Fig.Ā 3A, left panels). Likewise, the brains of IL-6ā/ā + rIL-6-infected mice exhibited dispersed GXM localization especially in the brainstem and prefrontal cortex (Fig.Ā 3A, right panels). In all the groups, there was significant GXM deposition surrounding cryptococcomas or areas of encephalomalacia. Analysis of the intensity of GXM staining in brain tissue images from Cn Wild-type, IL-6ā/ā, and IL-6ā/ā + rIL-6-infected mice revealed that animals supplemented with IL-6 had significantly lower GXM intensity or accumulation in all the brain regions analyzed than the IL-6ā/ā-group (Pā<ā0.0001; Fig.Ā 3B-F). Moreover, brains removed from IL-6ā/ā + rIL-6 brains had significantly higher GXM intensity than those excised from Wild-type mice (hippocampus and hypothalamus, Pā<ā0.0001; cortex and midbrain, Pā<ā0.001; cerebellum, Pā<ā0.05; Fig.Ā 3B-F). To validate that Wild-type mice naturally produce IL-6 in the brain during cryptococcal infection and that this cytokine has a crucial role in reducing GXM tissue accumulation, we measured IL-6 levels in homogenates from brains excised at 3- and 7-dpi (SFig.Ā 2). Wild-type-infected mice demonstrated high IL-6 production in brain tissue at 3 (Pā<ā0.01)- and 7 (Pā<ā0.0001)-dpi relative to Wild-type uninfected mice (nā=ā3 mice/group/day), with a 2-fold increase at 7-dpi over 3-dpi (Pā<ā0.001) or as the systemic infection progressed. Hence, our data show that IL-6 reduces fungal capsular polysaccharide brain accumulation, which may have important implications impacting the progression of cerebral cryptococcosis.
rIL-6 increased Cn capsule growth in vitro
The polysaccharide capsule is the main virulence factor of Cn. The extensive secretion of the capsule, and particularly GXM, compromises the immune responses of the host to combat the infection. To confirm our results indicating that IL-6 deficiency promotes Cn GXM release in brain tissue, we measured the effect of rIL-6 on capsular size and volume in vitro using immunofluorescence, India ink staining, and light/confocal microscopy (Fig.Ā 4). Specific IL-6 MAb was used to neutralize the effects of this pro-inflammatory cytokine on Cn capsular synthesis. Visual inspection of fluorescent images taken from cryptococci cultured in absence (untreated) or presence of rIL-6 (10Ā Āµg/mL) or rIL-6ā+āanti-IL-6 (20Ā Āµg/mL) for 48Ā h at 37Ā Ā°C revealed that this immunomodulator increases Cn capsule volume (blue halos; Fig.Ā 4A). Quantification of India ink-stained microscopic images (Fig.Ā 4A insets; nā=ā50 cells per group) confirmed that treatment with rIL-6 (454.3 Āµm3) significantly stimulated Cn capsular volume compared to untreated (279 Āµm3; Pā<ā0.0001) and rIL-6ā+āanti-IL-6 treatment (347.6 Āµm3; Pā<ā0.0001; Fig.Ā 4B). Also, rIL-6ā+āanti-IL-6-exposed cryptococci evinced higher capsule size than untreated fungal cells (Pā<ā0.01). Furthermore, to understand how rIL-6 affects Cn capsule formation in vitro, we performed quantitative polymerase chain reaction (qPCR) to determine the expression of Cap59 (Fig.Ā 4C) and Grasp (Golgi reassembly and stacking protein; Fig.Ā 4D), which are involved in capsular synthesis [26] and secretion [27], respectively. Culture of Cn with rIL-6 for 24Ā h significantly induced the expression of Cap59 and Grasp relative to untreated (Pā<ā0.05 for both) and rIL-6ā+āanti-IL-6 (Pā<ā0.05 for both). Although increasing (2 vs. 24Ā h; Cap59) and decreasing (2 vs. 24Ā h; untreated and rIL-6ā+āanti-IL-6; Grasp) gene expression trends were observed, these tendencies were not statistically significant (Fig.Ā 4C-D). To assure that the differences in capsule size and related gene expression were not due to variations in fungal growth, we monitored the proliferation of untreated and rIL-6- or rIL-6ā+āanti-IL-6-treated cryptococci in real time using Bioscreen C analysis (Fig.Ā 4E). Fungal cells in each condition exhibited similar growth for 24Ā h. Our results demonstrate that IL-6 promotes Cn capsule growth, and this effect is independent from cell replication, which may be a potential fungal defense mechanism in response to this host immunomodulator.
IL-6 deficiency promotes the presence of dystrophic microglia during Cn infection
Microglia are the resident phagocytes of the CNS, involved in surveillance and immune recruitment, and important inducers of inflammatory responses through cytokine signaling. We recently described that Cn infection modifies microglial morphology from branched or ramified (cylindrical soma with long and thin ramifications) to either activated or hypertrophic (thick soma and thick ramifications), phagocytic or amoeboid (large soma and short retracted ramifications; ameboid or macrophage-like), dystrophic (not-well defined soma and thin ramifications), or rod-shaped (enlarged cylindrical soma and polar ramifications) phenotypes, which may influence the progression and outcome of cerebral cryptococcosis [28]. IL-6 has been shown to stimulate brain repair by microglia particularly in traumatic brain injury [29]. Hence, we examined how IL-6 deficiency alters the morphology of microglia during cryptococcal infection at 3- and 7-dpi (Fig.Ā 5). Microglia were immunolabeled using ionized calcium-binding adaptor molecule-1 (Iba-1; brown)-binding MAb (Fig.Ā 5A and C). Images of each group treatment were visualized under the microscope (Fig.Ā 5A and C) and the morphology of microglia (Fig.Ā 5B and D) was qualitatively documented as previously described [28, 30]. Microglia were classified as activated or hypertrophic, dystrophic, phagocytic, or amoeboid, ramified, and rod-shaped [28].
At 3-dpi, the brain of a Wild-type mouse (10X, top left panel) shows a cryptococcal lesion surrounded by Iba-1+ cells or microglia (Fig.Ā 5A). High magnification (40X, center and 100X, bottom left panels) of Wild-type tissue demonstrate dystrophic microglia (red arrows) near the area of encephalomalacia consisting of a small cell body with a distorted surface and loss of their processes (purple arrowheads). IL-6ā/ā-infected brain tissue displayed ramified microglia (40X, center middle panel) which is characterized by having a relatively small ovoid cell body surrounded by very thin branches (orange arrow; 100X, bottom middle panel). The brain of an IL-6ā/ā + rIL-6-infected mouse exhibited increased microgliosis or increased number of microglia (10X, upper right panel). High magnification of IL-6ā/ā + rIL-6 brain tissue showed a cryptococcal lesion surrounded by activated and phagocytic microglia (black arrows; 40X, middle and 100X, bottom right panels).
Brain sections of IL-6ā/ā + rIL-6 had a significantly higher percentage of activated (42.5%) and phagocytic (35%) microglia than tissue from Wild-type (22.2%, activated; 19% phagocytic; Pā<ā0.05) and IL-6ā/ā (24.8%, activated; 9.2% phagocytic; Pā<ā0.05) animals (Fig.Ā 5B). IL-6ā/ā-infected brain tissue exhibited a significantly higher percentage of dystrophic (10.7%) microglia compared to Wild-type (4%; Pā<ā0.05) and IL-6ā/ā + rIL-6 (2.5%; Pā<ā0.05) mice. Likewise, brain tissue from IL-6ā/ā-infected mice showed a significantly higher percentage of branched or ramified (36.2%) microglia relative to the Wild-type (24.6%; Pā<ā0.05) and IL-6ā/ā + rIL-6 (9.2%; Pā<ā0.05) groups. Wild-type brains evinced a significantly higher percentage of ramified microglia than IL-6ā/ā mice (Pā<ā0.05). Lastly, rod-shaped (30.2%) microglia were the most abundant phenotype in infected Wild-type brain tissue at 3-dpi, with a higher percentage of this morphology relative to brain tissue from IL-6ā/ā (19.1%; Pā<ā0.05) and IL-6ā/ā + rIL-6 (10.8%; Pā<ā0.05). The percentage of rod-shaped microglia in IL-6ā/ā tissue was also significantly higher than in IL-6ā/ā + rIL-6 tissue (Pā<ā0.05).
The brains of Wild-type (10X, top left panel) and IL-6ā/ā + rIL-6 (10X, top right panel) mice excised at 7-dpi displayed increased microgliosis around the cryptococcomas (Fig.Ā 5C). In contrast, IL-6ā/ā brains exhibited less microglia surrounding the area of encephalomalacia (10X, top middle panel). Higher magnification of Wild-type brain tissue evinced dystrophic glial cells near the cryptococcoma (red arrows; 40X, center left panel) with close activated or hypertrophic microglia (black arrows; 40X, center and 100X, bottom left panels). IL-6ā/ā brains show dystrophic microglia (red arrows; 40X, center middle panel) and activated microglia that appears to be dropping their ramifications to become phagocytic (black arrows;Ā 100X, center bottom panel). IL-6ā/ā + rIL-6 brain tissue had considerable recruitment of phagocytic (blue arrows; 10X; top right panel) and activated (black arrows; 40X, right middle and 100X, bottom panels) microglia near the cryptococcal brain lesion. Notably, brain tissue from all the groups exhibited accumulation of microglial ramification or branch debris (purple arrow heads) that may have significant implications in Cn infection control.
IL-6ā/ā + rIL-6 brains had larger percentage of phagocytic microglia (40.9%) than Wild-type (29.7%; Pā<ā0.05) and IL-6ā/ā (19.2%; Pā<ā0.05) brains at 7-dpi (Fig.Ā 5D). In contrast, the brains of IL-6ā/ā mice showed a higher percentage of dystrophic cells (30.8%) than Wild-type (12.3%; Pā<ā0.05) and IL-6ā/ā + rIL-6 (4.7%; Pā<ā0.05) groups. All the groups have proportionally similar activated or hypertrophic microglia. Wild-type brains have the largest percentage of rod-shaped (22.5%; Pā<ā0.05) cells. Branched or ramified microglia was the least observed phenotype found in the brains of the compared groups, although Wild-type (7.2%) and IL-6ā/ā + rIL-6 (7.8%) mice had similar percentage of this morphology, whereas IL-6ā/ā mice had the lowest percentage (2.2%; Pā<ā0.05).
Taken together, our in vivo data suggest that IL-6 prevents brain colonization, potentially through its effects on microglia morphology and their effector functions.
NR-9460 microglia-like cells treated with IL-6 small interfering RNA (siRNA) exhibited decreased cryptococcal phagocytosis
Phagocytosis is one of the main activities associated with microglial responses to infection. Therefore, we assessed the impact of IL-6 on fungal phagocytosis by NR-9460 microglia-like cells. For that, we first transfected NR-9460 cells with IL-6 siRNA for 24Ā h to silence this cytokine gene and inhibit protein production. We similarly treated NR-9460 cells with a mock siRNA molecule and used these cells as a negative control. To ensure that siRNA treatments did not affect the viability of NR-9460 microglia-like cells, we performed flow cytometry analysis (nā=ā4 replicates per group) and compared siRNA groups to untreated cells (SFig.Ā 3). Representative flow cytometry dot plots are shown (SFig.Ā 3Ā A). On average, untreated (91.9%), siRNA IL-6 (88.8%), and siRNA negative control (84.4%) NR-9460 cells show similar cell viability percentage. Then, we activated untreated, siRNA IL-6, and siRNA negative control NR-9460 cells with bacterial lipopolysaccharide (LPS; 0.5Ā Āµg/mL)/IFN-Ī³ (5 ng/mL) in Opti-MEM I medium with 2% fetal bovine serum (FBS) for 2Ā h and collected their culture supernatant after 24Ā h incubation at 37Ā Ā°C and 5% CO2 (Fig.Ā 6A). Transfected cells with siRNA IL-6 evinced a significant 9 to 10-fold reduction in IL-6 production compared to untreated (Pā<ā0.0001) and siRNA negative control (Pā<ā0.0001) cells. There was no difference in IL-6 production between untreated and siRNA negative control cells. We showed the flow cytometry gating strategy utilized to quantify cryptococcal phagocytosis and representative dot plots for each condition (Fig.Ā 6B). siRNA IL-6 (81%)-treated NR-9460 microglia-like cells had significantly lower fungal phagocytosis percentage than untreated (84.5%; Pā<ā0.01) and siRNA negative control (84.1%; Pā<ā0.01) cells (Fig.Ā 6C). Our findings suggest that IL-6 production by microglia, although marginally, enhances cryptococcal phagocytosis, which is important for infection control especially upon brain penetration.
rIL-6 enhances cryptococcal phagocytosis and killing by microglia in vitro
We investigated the role of rIL-6 on the efficacy of GXM-specific MAb 18B7-mediated phagocytosis and killing of Cn by NR-9460 microglia-like cells after 4Ā h at 37Ā Ā°C and 5% CO2 (Fig.Ā 7). The gating strategy used in the flow cytometry analysis to quantify cryptococcal phagocytosis by NR-9460 cells as well as the representative dot plots for each experimental condition are shown (Fig.Ā 7A). NR-9460 cells cultured with rIL-6 demonstrated higher phagocytosis of cryptococci (25.1%) compared to untreated (14.13%; Pā<ā0.0001) and rIL-6ā+āanti-IL-6 (20.3%; Pā<ā0.01) microglia-like cells (Fig.Ā 7B). rIL-6ā+āanti-IL-6-treated microglia phagocytized more fungal cells than the untreated counterparts (Pā<ā0.001). In addition, microglial cells incubated with rIL-6 evinced higher cryptococcal killing than untreated (Pā<ā0.05) and rIL-6ā+āanti-IL-6-treated cells (Pā<ā0.05; Fig.Ā 7C). There was no difference in Cn killing between untreated and rIL-6ā+āanti-IL-6-treated microglia. These results indicate that IL-6 stimulates microglia to engulf and kill Cn cells, thus impacting the capacity of these CNS myeloid cells to control the infection and disease progression.
Cn infection induces only minor astrocytic responses in IL-6ā/ā mice
Cn causes astrocytes to become reactive during infection [13, 14] and these morphological alterations probably have a critical role in cryptococcal CNS colonization and meningoencephalitis development. Therefore, we assessed the astrocytic responses and morphological changes in brain tissue of Wild-type, -IL-6ā/ā, and -IL-6ā/ā + rIL-6 following systemic cryptococcal infection at 3- and 7-dpi (Fig.Ā 8). Astrocytes were immunolabeled with glial fibrillary acidic protein (GFAP; brown)-binding MAb. At 3-dpi, images of Wild-type brains exhibited astrocytosis with protoplasmic astroglia surrounding neurons and fibrous astrocytic subtypes surrounding the wall of blood capillaries in the white matter (10X, top left panel; Fig.Ā 8A). High magnification images (40 and 100X) of Wild-type brains displayed astrogliosis in the hippocampal parenchyma. Low (10X, center top panel) and high (40-100X, center middle and bottom panels) magnification images of IL-6ā/ā hippocampal tissue showed normal astrocytes with thin processes and without any reactive morphological changes. Finally, the hippocampus of IL-6ā/ā + rIL-6 mice exhibited considerable astrocytosis and astrogliosis (10-40X, top and middle right panels; Fig.Ā 8A). High magnification images (100X, bottom right panel) of IL-6ā/ā + rIL-6 hippocampal tissue displayed astrocytes with long thick-branched processes forming an arborizing pattern around neurons and blood vessels. To quantify the morphology of astrocytes in brains excised from Wild-type, IL-6ā/ā, and IL-6ā/ā + rIL-6-infected mice at 3-dpi, we used light microscopy to measure their processes number and thickness (Fig.Ā 8B-C). Wild-type- and IL-6ā/ā + rIL-6-astrocytes displayed significantly higher number of processes (Pā<ā0.001 and Pā<ā0.0001, respectively; Fig.Ā 8B) and thicker processes (Pā<ā0.0001 and Pā<ā0.001, respectively; Fig.Ā 8C) relative to IL-6ā/ā-derived astrocytes. No differences in astrocyte morphology were observed between Wild-type- and IL-6ā/ā + rIL-6 groups.
At 7-dpi, images of Wild-type brains displayed an increased number of brown-stained protoplasmic astrocytes in the hippocampus (10X, top left panel; Fig.Ā 8D). Interestingly, astrogliosis was observed (black arrow; 40X, center left panel) and the processes of an astrocyte were shown surrounding a blood vessel (black arrow; 100X, bottom left panel). IL-6ā/ā hippocampal tissue demonstrates minimal astrogliosis (10X, top middle panel) and long and thin astrocytic processes (black arrows; 40-100X; Fig.Ā 8D). Lastly, the hippocampus of IL-6ā/ā + rIL-6 exhibited moderate astrocytosis and astrogliosis (10X, top right panel; Fig.Ā 8D) with accumulation of astrocytes with intermediate process thickness (100X, bottom right panel). We also quantified the morphology of astrocytes in brains excised from Wild-type, IL-6ā/ā, and IL-6ā/ā + rIL-6-infected mice at 7-dpi (Fig.Ā 8E-F). Wild-type- and IL-6ā/ā + rIL-6-astrocytes displayed significantly higher number of processes (Pā<ā0.0001 for both; Fig.Ā 8E) and thicker processes (Pā<ā0.0001 for both; Fig.Ā 8F) relative to IL-6ā/ā-derived astrocytes. No differences in astroglial morphology were observed between Wild-type- and IL-6ā/ā + rIL-6 groups.
These findings show that brains removed from IL-6ā/ā mice have a weaker astrocytic response than Wild-type- or IL-6ā/ā + rIL-6 and that this response impacts considerably the ability of the immune response to control cryptococcal CNS infection and clearance.
Discussion
Using an IL-6 knock-out (KO) mouse model of systemic Cn infection, we demonstrated and validated the importance of this multifunctional cytokine on disseminated cryptococcosis, particularly focusing on fungal brain invasion and colonization. However, mice deficient in IL-6 had more pulmonary inflammation than Wild-type and IL-6ā/ā + rIL-6 mice at 3-dpi even when IL-6ā/ā + rIL-6 lungs had higher fungal load than those from Wild-type and IL-6ā/ā mice. Since Wild-type mice actively produce IL-6 early during cryptococcal infection, these animals were able to limit the inflammatory response to localized regions of the lungs while keeping the fungal burden lower than IL-6ā/ā and IL-6ā/ā + rIL-6 groups. As the infection progressed, mice deficient in IL-6 had similar histological respiratory disease development compared to animals with intact IL-6 responses or supplemented with the exogenous cytokine. The lungs of mice in each experimental group displayed considerable inflammation, similar cryptococcoma formation, and comparable fungal burden at 7-dpi, suggesting that IL-6 production may only play a critical role early in cryptococcal pulmonary disease, or its function can be substituted by other cytokines involved in the acute phase response such as IL-1Ī² and TNF-Ī±.
IL-6ā/ā mice exhibited faster mortality than animals that naturally produced or were supplemented with the cytokine likely due to their inability to control fungal replication in circulation or penetration into the CNS. In this regard, IL-6 has been shown to contribute to the inhibition of cryptococcal proliferation [31], which may explain the reduced fungal load in the blood of Wild-type and IL-6ā/ā + rIL-6 mice. Also, IL-6ā/ā mice have been shown to increase sepsis severity and mortality in a model of cecal ligation and puncture [32], which may explain why IL-6ā/ā + rIL-6 mice evinced comparable high number of cryptococci in blood to IL-6ā/ā animals at 3-dpi. Nevertheless, consistent rIL-6 administration to IL-6-deficient mice controlled the fungal load in circulation to equivalent numbers to those observed in Wild-type mice at 7-dpi. Low IL-6 levels in CSF are correlated with reduced survival in patients with HIV/AIDS and cryptococcal meningoencephalitis [33]. IL-6 deficiency has been previously shown to compromise the BBB integrity [22], facilitating Cn brain invasion, which explains the increased fungal burden in these mice compared to those with IL-6. IL-6 neutralization after specific MAb administration to C57BL/6 mice has been shown to increase the BBB permeability of Cn [22]. We also confirmed that injection of IL-6 to KO mice reduces cryptococcal invasion into the CNS likely by strengthening the integrity of the BBB [22]. Moreover, resistance to Cn in the brains of mice correlated with the local production of IL-6 and IL-1Ī², and that resistance increases by the addition of either one exogenously before fungal challenge [24]. These previous observations also support our findings showing that IL-6ā/ā brains had higher fungal burden than Wild-type and IL-6ā/ā + rIL-6 mice.
Brain tissue removed from IL-6ā/ā mice had substantial accumulation of Cn GXM. GXM aggregation reduces inflammation [33] and prevents immune cell infiltration into the brain during disseminated infection [34]. We recently demonstrated that GXM disrupts endothelial cell tight junctions in the BBB, facilitating the transmigration of Cn into the CNS [3]. In contrast, Wild-type mice showed significantly lower GXM intensity in the brain than IL-6ā/ā and IL-6ā/ā + rIL-6 mice. To rule out that the reduction of GXM accumulation observed in Wild-type mice was not attributed to being unable to actively producing IL-6 in infected brain tissue, we compared uninfected vs. infected Wild-type mice and confirmed that Wild-type-infected mice produced IL-6 in brain tissue and that its concentrations increased as the infection progressed. Beenhouwer et al., previously demonstrated that Wild-type mice had lower GXM in serum than IL-6 KO mice [35]. Although we did not measure the levels of GXM in circulation (e.g., serum or CSF), high GXM levels in the blood of HIV/AIDS patients with cryptococcosis are associated with lower IgG production and mortality [36]. Secreted GXM accumulates in patient serum and CSF at Āµg/mL concentrations and has well-documented immunosuppressive properties, correlating with poor patient outcomes. For example, we recently demonstrated in intracerebrally infected mice that mortality was associated with the presence of subarachnoid hemorrhaging and GXM deposition in the meningeal blood vessels and meninges in all brain regions infected [12]. Wild-type and IL-6ā/ā + rIL-6 brains exhibited lower GXM intensity, validating that IL-6 is critical and beneficial for mouse survival by attenuating the progression of neurocryptococcosis [24]. Importantly, the administration of rIL-6 is artificial, compared to what occurs in infected Wild-type mice, thus, explaining the higher GXM tissue accumulation observed in IL-6ā/ā + rIL-6 mice and vice versa when compared to IL-6ā/ā mice.
Cn can adaptively modulate the size and polysaccharide release of its capsule according to external stimuli, which can be advantageous to the fungus during pathogenesis. Exposure of cryptococci to rIL-6 induced the expression of capsular-related genes Cap59 and Grasp and significantly stimulated the enlargement of the capsule (Fig.Ā 9A) relative to untreated and IL-6ā+āanti-IL-6 cells. Also, rIL-6-mediated capsule size augmentation by Cn was independent of fungal proliferation since cryptococci grew similarly in each condition in vitro. Activation of peripheral blood mononuclear cells after interactions with Cn increases the levels of IL-6 and augments their resistance to infection [31]. Clinical cryptococcal strains producing larger ex vivo capsules in the baseline CSF correlated with higher intracranial pressure, slower fungal clearance, and paucity of CSF inflammation, including decreased CSF white blood cell count, IL-4, IL-6, IL-7, IL-8, and IFN-Ī³ [37]. IL-6 is produced by neutrophils in response to Cn capsule size via interaction of GXM with complement [38]. It is possible that even when IL-6 enhances Cn capsule size, infected Wild-type mice were able to control the fungal burden in blood and brain because host phagocytes likely increased their Ab-mediated fungal phagocytic activity [39]. The clearance of Cn overlaps with the development of an adaptive immune response that enhances killing or containment of the fungi in granulomas [40]. Therefore, successful containment of the fungus requires both innate and adaptive immune responses. Interestingly, deletion of the Apt1 flippase in Cn reduces the packaging of GXM into extracellular vesicles and impairs GXM synthesis whereas it reduces the production of IL-6 during infection and CNS colonization [41]. The capsule size of IL-6ā+āanti-IL-6 cells were larger than those of untreated cryptococci, suggesting that the modulation of the capsule may be triggered by sensing or interacting with either the cytokine, Ab, or their complex.
We found that IL-6 deficiency resulted in different microglial morphology phenotypes in response to Cn infection particularly characterized by an abundance of dystrophic cells. The accumulation of dystrophic microglia is associated with tissue degeneration [42] or senescence [43]. Aging individuals with Down syndrome [43] and Alzheimerās disease [44] present an increased number of dystrophic microglia, which are unable to carry out their homeostatic functions [43]. In fact, dystrophic microglia rather than activated microglia are present with tau pathology and may precede neurodegeneration in Alzheimerās disease [44]. It is possible that the identification of dystrophic microglia in Cn-infected brain tissue may be a marker for the progression stage of cerebral cryptococcosis or cognitive decline in patients that recovered from the infection. In contrast, Wild-type and IL-6ā/ā + rIL-6 brains had higher number of activated and phagocytic microglia in infected tissue, with these cell phenotypes surrounding the cryptococcomas or brain lesions, which are associated with stronger response to infection and tissue repair. It is important to highlight that of the different microglial phenotypes evaluated, IL-6ā/ā + rIL-6 brains hadāā„ā75% of activated and phagocytic microglia throughout the infection and may explain the longer survival of these mice even though they showed high fungal burden early. While Wild-type and IL-6ā/ā brains exhibited a high percentage of ramified microglia early during the infection. For example, microglia cultured with rIL-6 or even neutralized rIL-6 demonstrated more phagocytosis and fungal killing than untreated cells (Fig.Ā 9B). IL-6, in addition to IL-1Ī², TNF-Ī±, and IL-12, is produced by microglia-like cells during interactions with acapsular or encapsulated strains of Cn [45]. Thus, it is plausible that IL-12 [46] and IL-6 [47] production by microglia stimulate a combination of T helper cells 1 and 2 (Th1 or 2), respectively, to facilitate engulfing of the yeast cells (Th2) and enhance killing of the fungus in the phagolysosome (Th1). Moreover, silencing IL-6 expression in microglia-like cells reduced phagocytosis of Cn cells after activation with LPS/IFN-Ī³ (Fig.Ā 9C). In addition to combat infection, microglia can be neuroprotective and may promote brain tissue repair in an IL-6-dependent manner [29] to alleviate the cognitive deficits arising from tissue injury in recovering patients from cryptococcal meningoencephalitis, although further studies are required to test this hypothesis.
We identified extensive microglia cell body debris in brain tissue from each experimental group, particularly in the boundaries of the regions with encephalomalacia. It is provocative to postulate that extensive Cn GXM secretion and its intimate interaction with microglia may cause the separation of these microglia cell body ramifications, which can accumulate in tissue and compromise the inflammatory responses. In this regard, HIV/AIDS patients with cryptococcal meningoencephalitis typically exhibit minimal inflammation due to their reduced number of CD4+ T cells [14]. Another possibility is that GXM induces the transition of microglia from ramified to dystrophic, compromising their effector functions. Future studies are necessary to validate these hypotheses and expand our insight on microglial responses to Cn CNS invasion and colonization.
Substantial astrocytosis and astrogliosis were observed in Cn-infected tissue slices from Wild-type and IL-6ā/ā + rIL-6 mice. Considerable astrocyte reactivity was evident in Wild-type brain tissue and these cells produce high levels of IL-6, especially in traumatic brain injury [48]. Reactive astrocytes are found nearby large brain lesions or overlapping with fungal extracellular vesicles in human post-mortem tissue from individuals with HIV/AIDS [14] and mice [49], respectively, likely playing a critical and understudied role in containing infection [50]. Human astrocytes inhibit cryptococcal proliferation through activation of NO [51] and recently we showed that astrocyte NO activation is higher in astroglia than microglia [28]. Calcium binding-S100B protein is secreted by astrocytes and stimulates NO secretion by these glial cells in an autocrine manner [17]. Nevertheless, Cn can neutralize astrocyte derived NO without interfering with inducible NO synthase generation or catalytic activity [52]. We observed minimal astrogliosis in IL-6ā/ā brain tissue infected with Cn; however, this was not surprising because these mice have previously shown impaired astroglia activation [53], indicating the importance of IL-6 in stimulating glia responses to fight this CNS mycosis.
Although studies by Li [22] and Blasi [24] had previously demonstrated the critical role of IL-6 in combating cerebral cryptococcosis, our study is unique relative to those seminal publications in that we used a validated clinical and highly virulent Cn (strain H99; serotype A; var. grubii) isolate. Cn invades the CNS in higher rates than C. deneoformans (serotype D; var. neoformans) and C. gattii (serotype B/C). Li and colleagues [22] used a C. deneoformans strain from unknown origin and low virulence, which explains why using a high inoculum (3.6āĆā106 CFU) to infect mice intranasally (i.n.) resulted in 90% survival rate of Wild-type C57BL/6 male mice and 100% mortality in IL-6ā/ā mice after 120-dpi. In fact, C57BL/6 mice infected i.n. with 105Cn H99 cells (a considerably lower inoculum) caused 100% mortality at 32-dpi [54], highlighting the importance of understanding the pros and cons of each study when making data analysis and interpretation. Another important discrepancy is that even though the IL-6ā/ā mice in the Liās study showed increased brain fungal burden as the disease progressed, the Wild-type mice had minimal cryptococcal brain invasion throughout the duration of the infection [22]. This is interesting because Coelho and colleagues have shown that H99 cryptococci rapidly (<ā3Ā h) invades the brain after nasal infection [55]. Similarly, Blasi and colleagues [24] used the Cn (Sanfelice) Vuillemin (ATCC 11240) or Cn var. shanghaiensis Liao (described in 1980s) strain, an environmental isolate that is now classified as C. gattii [56]. C. gattii became prominent after a cryptococcosis outbreak on Vancouver Island, British Columbia, Canada particularly affecting individuals with apparent immunocompetency [57]. Although C. gattii causes pulmonary and CNS infection, Cn is still isolated from and associated with most of the cases of cryptococcosis (42.4% vs. 1.3%) in endemic regions such as sub-Saharan Africa [58]. Blasi also showed that exogenous and local (intracerebrally; i.c.) IL-6 supplementation of Wild-type mice prior to i.c. cryptococcal infection significantly reduced blood and brain fungal load, resulting in prolonged survival. Despite our study showing a similar result on the impact of IL-6 delaying mouse mortality, our findings are novel in that we not only corroborated the work by Li et al.. and Blasi et al., but further their observations by showing the importance of this cytokine on IL-6ā/ā mice after exogenous systemic supplementation and on Cn by molecularly modulating its capsular production. Furthermore, this is the first time a specific study has been done on the effects IL-6 has on microglia and astrocyte responses to cerebral cryptococcosis in vivo, which is an important step forward to understanding the way Cn establishes and develops an infection in the brain environment.
In conclusion, we have demonstrated that IL-6 has a significant role in combating Cn systemic infection in mice. The molecular mechanisms underlying this effect remain unknown. Our data suggest that IL-6 deficiency causes early inflammation but does not alter pulmonary disease as the infection progresses. IL-6 deficiency enhances cryptococcal proliferation in the bloodstream, which could potentially facilitate fungal crossing into the CNS, leading to alterations in glial activation and responses that are associated with increased mortality (Fig.Ā 10). Further studies investigating the impact of immunomodulator molecules such as IL-6 on cerebral cryptococcosis are warranted for the development of antifungal therapy or treatments aimed to prevent, reduce, or manage Cn infections especially in at-risk immunosuppressed populations.
Materials and methods
Cn
Cn strain H99 (serotype A) was isolated and kindly provided by John Perfect at Duke University. Yeasts were grown in Sabouraud dextrose broth (pH 5.6; BD Difco) for 24Ā h at 30Ā Ā°C in an orbital shaker (New Brunswick Scientific) set at 150Ā rpm (to early stationary phase). They were washed, counted, and resuspended in sterile 0.9% sodium chloride (saline) solution, USP (Baxter).
NR-9460 cells
The murine microglial cell line NR-9460 (BEI Resources, NIAID, NIH) was derived from wild-type mouse brain tissue and immortalized by infection with the ecotropic transforming replication-deficient retrovirus J2. Characterization based on immunofluorescence, stimulation assays, and flow cytometry demonstrated that the NR-9460 cell line retains its glia-specific morphological, functional, and surface expression properties.
Systemic infection and rIL-6 administration
Female C57BL/6 and B6.129S2-Il6tm1Kopf/J mice (IL-6ā/ā; 6 to 8 weeks old; 20 to 25Ā g; Jackson Laboratory) were injected in their tail vein with a 100-ĀµL suspension containing 105Cn H99 cells. The intravenous (IV) injection was selected due to its high reproducibility in Cn CNS infection models [28]. To mimic baseline physiological IL-6 levels [59], one-day prior to infection, a group of IL-6ā/ā mice were injected IP with rIL-6 (1.6 pg/g) diluted in sterile saline solution. One-dpi, mice were injected with a 100-ĀµL solution of sterile saline (untreated and IL-6ā/ā) or supplemented with rIL-6 (40 pg/g/day; [32]) IP every other day. The daily dose of rIL-6 was selected and calculated accordingly based on a previous report on cryptococcal infection that showed IL-6 serum concentrations of 500 pg/mL/mouse [60]. Animals were monitored for survivability or pathology (Fig.Ā 1). The survival end points included inactivity, tachypnea, or loss of ā„ā25% of body weight from baseline weight. We monitored the mice twice daily for clinical signs (e.g., temperature), dehydration, and weight loss. Animals showing signs of dehydration or that lost more than 10% body weight received supportive care such as 1 mL of parenteral fluid supplementation (saline) and moist chow on the cage floor was provided.
CFU determinations
Mice were bled from their facial vein using heparinized tubes for collection and euthanized at 3- and 7-dpi. Rapidly, lungs and brain were excised and weighed. The right-lung lobe and left-brain hemisphere tissues were homogenized in 5 mL of sterile phosphate-buffered saline (PBS, pH 7.3āĀ±ā0.1). Blood and tissue homogenates were serially diluted, and a 100-ĀµL suspension was plated on Sabouraud dextrose agar (BD Difco) and incubated at 30Ā Ā°C for 48Ā h. Quantification of viable yeast cells from infected animals (nā=ā3ā5/group/day) was determined by CFU counting of two dilutions per animal (nā=ā6 plates per fluid or tissue).
Histopathology
The left-lung lobe and right-brain hemisphere were harvested and immersed in 4% paraformaldehyde (Fisher) overnight instead of perfusing the organs. Even though inflation of the lungs via intratracheal instillation of fixative is recommended to best preserve lung morphology and reduce artifactual atelectasis, this approach is contraindicated for lung infection as this can alter the anatomic location of fungal cells and cellular debris [61]. Then, tissues were washed 3X with sterile saline for 1Ā h, embedded in paraffin, 4Ā Ī¼m coronal sections were serially cut using a cryostat (Tanner Scientific, model: TN50), fixed onto glass slides, and subjected to hematoxylin-eosin (lung) or mucicarmine (brain) staining (nā=ā3 mice per group) to examine host tissue or fungal morphology, respectively. GXM (MAb 18B7 is an anti-cryptococcal GXM IgG1 generated and generously provided by Arturo Casadevall at the Johns Hopkins Bloomberg School of Public Health; 1:1,000 dilution), Iba-1 (rabbit anti-human Iba-1; 1:1,000 dilution; FujiFilm Wako), and GFAP (rabbit anti-human GFAP 2033X; 1:2,000 dilution; Dako) specific Ab (conjugated to horseradish peroxidase; dilution: 1:1,000; Santa Cruz Biotechnology) immunostaining to assess capsular release, microglial phenotype, and astrocyte morphology, respectively, near cryptococcomas. The slides were visualized blindly by three independent investigators using a Leica DMi8 inverted microscope, and images were captured with a Leica DFC7000 digital camera using LAS X digital imaging software. The morphology of microglia and astrocytes in brain tissue was quantified (nā=ā3 mice per group) by LAS X digital imaging software using the recorded 40X images and standardized 250āĆā250-Āµm2 squares near cryptococcomas. GXM distribution in tissue sections at 10X magnification (nā=ā10ā15 fields per brain region; nā=ā3 mice per group) was calculated using NIH ImageJ color deconvolution tool software (version 1.53q). The mean color intensity of the GXM for each treatment group was plotted in Prism 10.1.2. (GraphPad). The images were also examined, analyzed, and described by Dr. Mohamed F. Hamed, a veterinary pathologist.
Brain IL-6 determinations
Brain tissue (0.2Ā g) was placed in 1.8 mL of RIPA buffer and homogenized, and the supernatant was stored at āā20Ā Ā°C until analyzed. An enzyme-linked immunosorbent assay (ELISA) was performed using the Preprotech kit for each cytokine following the manufacturerās protocol. Briefly, microtiter polystyrene plates were coated with capture anti-IL-6 (1Ā Āµg/mL) overnight and incubated at 4Ā Ā°C. Next day, each well was blocked with 1% bovine serum albumin in PBS for 2Ā h at room temperature (RT). Next, the brain samples were serially diluted on the plate using a multi-pipette and incubated overnight at 4Ā Ā°C. The ELISA was completed by adding 0.5Ā Āµg/mL of a specific detection Ab followed by a 2Ā h incubation at RT, then by avidin-horseradish peroxidase conjugate (HRP; 1:2,000 dilution) for 30Ā min at RT, and finally revealing by adding 2,2ā²-azinobis (3-ethylbenzothiazoline-6-sulfonic acid)-diammonium salt substrate. In each step, the wells were washed with 0.05% Tween 20 in PBS. The optical density was assessed at 450Ā nm using a BioTek Synergy LX Multimode Reader and monitored every 10Ā min for 1Ā h. Each sample was tested in triplicates in two independent ELISA measurements.
Confocal microscopy
For immunofluorescence studies, yeasts were incubated in 96-well microtiter plates with glass bottom for 1Ā h at 30Ā Ā°C with FUN-1 (green/red; 10 ĀµM; 470/590 nm; Invitrogen), washed 3 times with PBS, and incubated with MAb 18B7 (2Ā Āµg/mL) for 1Ā h at 37Ā ĀŗC, followed by allophycocyanin-conjugated goat anti-mouse IgG1 (blue; 651/660 nm; Thermo Fisher). Pictures were taken in a Nikon Eclipse Ti2 Inverted confocal microscope at 20X magnification and processed with NIS-Elements Imaging software.
Capsule measurements with India ink
The capsule volume of Cn cells cultured in the absence (untreated) or presence of rIL-6 or rIL-6ā+āanti-IL-6 for 24Ā h was measured. To observe and measure the size of the capsule, 10 ĀµL of each cell suspension were mixed with an India Ink (BD) drop and the capsules visualized under light microscopy. Images were randomly taken with a Leica DMi8 inverted microscope and DFC7000 T digital camera. The diameters of both capsule and cell body were measured using the Leica software platform LAS X. Capsule volume was calculated using the volume formula, where Rā=āradius of capsule and rā=āradius of cell body: capsule volume (V)ā=ā4/3 Ļ (R3 ā r3). Fifty cells were analyzed per condition.
RNA extraction and cDNA synthesis
RNA extraction was performed at 2 and 24Ā h after incubation of cryptococci in absence or presence of either rIL-6 or rIL-6ā+āanti-IL-6 using the Quick-RNA fungal/bacterial extraction kit (Zymo Research), following the manufacturerās instructions. To remove any genomic DNA carryover, the samples were treated with DNase I (Zymo Research) for 15Ā min at RT, followed by RNA recovery in DNase/RNase free water. Finally, 200 ng of total RNA were used to synthesize cDNA using the Verso cDNA Synthesis kit (Thermo Fisher), following the manufacturerās instructions. The control reaction was set up using all components of the reaction mixture but without RNA sample.
qPCR
The genes selected for quantification were Cap59 [26] and Grasp [27], both involved in capsule synthesis. The primers and annealing temperatures used for qPCR analysis are described in TableĀ 1. The expression of genes was determined by qPCR using PowerUp Syber Green Master Mix 2x (Applied Biosystems). Reactions were set up using 250 nM primers and 4.5 ĀµL of the cDNA template (diluted 1:10). The cycling conditions used were as follows: 50Ā Ā°C for 2Ā min, 95Ā Ā°C for 2Ā min, and then 40 amplification cycles of 95Ā Ā°C for 20Ā s, 52ā56Ā Ā°C for 30Ā s, and 72Ā Ā°C for 30Ā s. The samples were cooled to 55Ā Ā°C, and a melting curve for temperatures between 55 and 95Ā Ā°C, with 1Ā Ā°C increments, was recorded. Relative expression was determined using the 2-DDCT method on a qTower thermocycler (Analytik Jena). A non-template for qPCR was used as control and all reactions were carried out in triplicate. Target gene expression was measured using expression relative to that of the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) reference gene as well as the untreated cryptococci as reference sample.
Cn growth assessment
Fungal growth (106 cells) in minimal medium without (untreated) or with rIL-6 (10 ng/mL) or rIL-6 (10 ng/mL)ā+āanti-IL-6 (20 ng/mL) was assessed in real-time at an optical density (OD) of 600Ā nm every 30Ā min using a microplate reader (Bioscreen C; Growth Curves USA). Ten replicates were analyzed per condition.
Cell transfection assay
siRNA transfection of NR-9640 cells was performed with a pre-designed mouse siRNA IL-6 (Invitrogen) and a siRNA negative control (Invitrogen). One day prior transfection, NR-9640 cells were equally placed in 12- or 24-well plates at a confluency of 70% and incubated in Opti-MEM I reduced serum supplemented with 5% FBS at 37Ā°C and 5% CO2. On the day of the transfection, Opti-MEM I reduced serum medium without FBS was used to prepare the mix of the Lipofectamineā¢ RNAiMAX Transfection Reagent (Invitrogen) and each siRNA at a final concentration of 50 nM followed by an incubation at RT for 10 min. The mixture was then added to the microglia-like cells and incubated for 24Ā h at 37Ā°C and 5% CO2. Reverse transfection was also performed by preparing the complexes inside the wells and then adding cells and medium, according to the manufacturerās protocol. Transfection efficiency was detected after 24Ā h and an additional 2Ā h treatment with activation media [Opti-MEM I with 2% FBS and supplemented with LPS (0.5 Āµg/mL)/IFN-Ī³ (5 ng/mL) to stimulate the production of IL-6. Relative IL-6 expression was measured via qPCR using the following specific oligonucleotides: forward 5āCACGGCCTTCCCTACTTCAC3ā and reverse 5āTGCAAGTGCATCATCGTTGT3ā. RNA extraction and cDNA synthesis after siRNA treatment were performed using the Quick-RNA MiniPrep Plus (Zymo Research), following the manufacturerās instructions. To remove any genomic DNA carryover, the samples were treated with DNase I (Zymo Research) for 15Ā min at RT, followed by RNA recovery in DNase/RNase free water. Finally, 100 ng of total RNA were used to synthesize cDNA using the Verso cDNA Synthesis kit (Thermo Fisher), following the manufacturerās instructions. Two control reactions were set up using all components of the reaction mixture but without RNA sample or reverse transcriptase. The IL-6 protein levels were measured in the supernatant of NR-9460 cell culture 24Ā h after activation by ELISA as described. Activated NR-9460 cells with LPS/IFN-Ī³ without any siRNA but exposed to the Lipofectamine RNAiMAX Transfection Reagent were used as a reference of IL-6 expression/secretion.
Phagocytosis assay
Monolayers of NR-9640 cells were washed thrice with PBS, and Dulbeccoās Modified Eagle Medium (DMEM; feeding medium; [62, 63]) supplemented with IFN-Ī³ (5 ng/mL) and LPS (0.5Ā Āµg/mL) was added, followed by the addition of fluorescein isothiocyanate (FITC)-stained (1Ā h at RT, shaking) pre-opsonized cryptococci with MAb 18B7 (10Ā Āµg/mL) for 1Ā h, in a microglia : cryptococci ratio of 1:10 (5āĆā105 : 5āĆā106 cells). The plates were incubated for 4Ā h at 37Ā Ā°C and 5% CO2 for phagocytosis. For phagocytosis assessment, the monolayer coculture was washed thrice with PBS to remove nonadherent cells, trypsinized for 1Ā min to detach microglia, centrifugated, and resuspended in FACS buffer. Cells were strained through a 100Ā Ī¼m filter, stained with 7-aminoactinomycin D (7-AAD; Invitrogen) for viability, and fixed. Samples were processed on a BD Accuri C6 flow cytometer and Cn phagocytosis by microglia was analyzed using the FlowJo software. The percentage of phagocytosis per sample was determined by gating phagocytic microglia showing fluorescence at 516Ā nm after ā„ā10,000 events.
Killing assay
After Ab-mediated phagocytosis, each well containing interacting microglia-cryptococci was gently washed with feeding medium three times to get rid of fungal cells which were not phagocytized. Then, cryptococcus-engulfed microglia were incubated for 24Ā h at 37Ā Ā°C and 5% CO2. Microglia-like cells were lysed by forcibly pulling the culture through a 27-gauge needle 5 to 7 times. A 100-ĀµL volume of suspension containing cryptococci was aspirated from the wells and transferred to a microcentrifuge tube with 900 ĀµL PBS. For each well, serial dilutions were performed and plated in triplicate onto Sabouraud dextrose agar plates, which were incubated at 30Ā Ā°C for 48Ā h. Viable cryptococcal cells were quantified as CFU. Although it is plausible that noninternalized cryptococci could replicate for several generations in feeding medium during a 24-h period, wells for each condition were microscopically monitored after phagocytosis to reduce the possibility of obtaining confounding results.
Statistical analysis
All data were subjected to statistical analysis using Prism 10.1.3 (GraphPad). Differences in survival rates were analyzed by the log-rank test (Mantel-Cox). P values for multiple comparisons were calculated by one-way analysis of variance (ANOVA) and were adjusted by use of the Tukeyās post hoc analysis. P values of <ā0.05 were considered significant.
Data availability
No datasets were generated or analysed during the current study.
References
Neilson JB, Fromtling RA, Bulmer GS. Cryptococcus neoformans: size range of infectious particles from aerosolized soil. Infect Immun. 1977;17(3):634ā8.
Chang YC, Stins MF, McCaffery MJ, Miller GF, Pare DR, Dam T, Paul-Satyaseela M, Kim KS, Kwon-Chung KJ. Cryptococcal yeast cells invade the central nervous system via transcellular penetration of the blood-brain barrier. Infect Immun. 2004;72(9):4985ā95.
Lee HH, Carmichael DJ, Ribeiro V, Parisi DN, Munzen ME, Charles-Nino CL, Hamed MF, Kaur E, Mishra A, Patel J, et al. Glucuronoxylomannan intranasal challenge prior to Cryptococcus neoformans pulmonary infection enhances cerebral cryptococcosis in rodents. PLoS Pathog. 2023;19(4):e1010941.
Charlier C, Nielsen K, Daou S, Brigitte M, Chretien F, Dromer F. Evidence of a role for monocytes in dissemination and brain invasion by Cryptococcus neoformans. Infect Immun. 2009;77(1):120ā7.
Bicanic T, Muzoora C, Brouwer AE, Meintjes G, Longley N, Taseera K, Rebe K, Loyse A, Jarvis J, Bekker LG, et al. Independent association between rate of clearance of infection and clinical outcome of HIV-associated cryptococcal meningitis: analysis of a combined cohort of 262 patients. Clin Infect Dis. 2009;49(5):702ā9.
Rajasingham R, Govender NP, Jordan A, Loyse A, Shroufi A, Denning DW, Meya DB, Chiller TM, Boulware DR. The global burden of HIV-associated cryptococcal infection in adults in 2020: a modelling analysis. Lancet Infect Dis. 2022;22(12):1748ā55.
Fromtling RA, Shadomy HJ, Jacobson ES. Decreased virulence in stable, acapsular mutants of cryptococcus neoformans. Mycopathologia. 1982;79(1):23ā9.
Goldman DL, Lee SC, Casadevall A. Tissue localization of Cryptococcus neoformans Glucuronoxylomannan in the presence and absence of specific antibody. Infect Immun. 1995;63(9):3448ā53.
Vecchiarelli A. Immunoregulation by capsular components of Cryptococcus neoformans. Med Mycol. 2000;38(6):407ā17.
Pettoello-Mantovani M, Casadevall A, Smarnworawong P, Goldstein H. Enhancement of HIV type 1 infectivity in vitro by capsular polysaccharide of Cryptococcus neoformans and Haemophilus influenzae. AIDS Res Hum Retroviruses. 1994;10(9):1079ā87.
Goldman DL, Casadevall A, Cho Y, Lee SC. Cryptococcus neoformans meningitis in the rat. Lab Invest. 1996;75(6):759ā70.
Hamed MF, Enriquez V, Munzen ME, Charles-Nino CL, Mihu MR, Khoshbouei H, Alvina K, Martinez LR. Clinical and pathological characterization of Central Nervous System cryptococcosis in an experimental mouse model of stereotaxic intracerebral infection. PLoS Negl Trop Dis. 2023;17(1):e0011068.
Lee SC, Casadevall A, Dickson DW. Immunohistochemical localization of capsular polysaccharide antigen in the central nervous system cells in cryptococcal meningoencephalitis. Am J Pathol. 1996;148(4):1267ā74.
Lee SC, Dickson DW, Casadevall A. Pathology of cryptococcal meningoencephalitis: analysis of 27 patients with pathogenetic implications. Hum Pathol. 1996;27(8):839ā47.
Kozel TR, Highison B, Stratton CJ. Localization on encapsulated Cryptococcus neoformans of serum components opsonic for phagocytosis by macrophages and neutrophils. Infect Immun. 1984;43(2):574ā9.
Casadevall A. Antibody immunity and invasive fungal infections. Infect Immun. 1995;63(11):4211ā8.
Adami C, Sorci G, Blasi E, Agneletti AL, Bistoni F, Donato R. S100B expression in and effects on microglia. Glia. 2001;33(2):131ā42.
Papanicolaou DA, Wilder RL, Manolagas SC, Chrousos GP. The pathophysiologic roles of interleukin-6 in human disease. Ann Intern Med. 1998;128(2):127ā37.
Lyson K, McCann SM. The effect of interleukin-6 on pituitary hormone release in vivo and in vitro. Neuroendocrinology. 1991;54(3):262ā6.
Rothaug M, Becker-Pauly C, Rose-John S. The role of interleukin-6 signaling in nervous tissue. Biochim Biophys Acta. 2016;1863(6 Pt A):1218ā27.
Breen EC, Rezai AR, Nakajima K, Beall GN, Mitsuyasu RT, Hirano T, Kishimoto T, Martinez-Maza O. Infection with HIV is associated with elevated IL-6 levels and production. J Immunol. 1990;144(2):480ā4.
Li X, Liu G, Ma J, Zhou L, Zhang Q, Gao L. Lack of IL-6 increases blood-brain barrier permeability in fungal meningitis. J Biosci. 2015;40(1):7ā12.
Lortholary O, Improvisi L, Rayhane N, Gray F, Fitting C, Cavaillon JM, Dromer F. Cytokine profiles of AIDS patients are similar to those of mice with disseminated Cryptococcus neoformans infection. Infect Immun. 1999;67(12):6314ā20.
Blasi E, Barluzzi R, Mazzolla R, Pitzurra L, Puliti M, Saleppico S, Bistoni F. Biomolecular events involved in anticryptococcal resistance in the brain. Infect Immun. 1995;63(4):1218ā22.
Redlich S, Ribes S, Schutze S, Eiffert H, Nau R. Toll-like receptor stimulation increases phagocytosis of Cryptococcus neoformans by microglial cells. J Neuroinflammation. 2013;10:71.
Chang YC, Kwon-Chung KJ. Complementation of a capsule-deficient mutation of Cryptococcus neoformans restores its virulence. Mol Cell Biol. 1994;14(7):4912ā9.
Kmetzsch L, Joffe LS, Staats CC, de Oliveira DL, Fonseca FL, Cordero RJ, Casadevall A, Nimrichter L, Schrank A, Vainstein MH, et al. Role for golgi reassembly and stacking protein (GRASP) in polysaccharide secretion and fungal virulence. Mol Microbiol. 2011;81(1):206ā18.
Hamed MF, Araujo GRS, Munzen ME, Reguera-Gomez M, Epstein C, Lee HH, Frases S, Martinez LR. Phospholipase B is critical for Cryptococcus neoformans Survival in the Central Nervous System. mBio. 2023;14(2):e0264022.
Willis EF, MacDonald KPA, Nguyen QH, Garrido AL, Gillespie ER, Harley SBR, Bartlett PF, Schroder WA, Yates AG, Anthony DC, et al. Repopulating Microglia promote Brain Repair in an IL-6-Dependent manner. Cell. 2020;180(5):833āe846816.
Munzen ME, Reguera Gomez M, Hamed MF, Enriquez V, Charles-Nino CL, Dores MR, Alvina K, Martinez LR. Palmitoylethanolamide shows limited efficacy in controlling cerebral cryptococcosis in vivo. Antimicrob Agents Chemother. 2023;67(10):e0045923.
Siddiqui AA, Shattock RJ, Harrison TS. Role of capsule and interleukin-6 in long-term immune control of Cryptococcus neoformans infection by specifically activated human peripheral blood mononuclear cells. Infect Immun. 2006;74(9):5302ā10.
Remick DG, Bolgos G, Copeland S, Siddiqui J. Role of interleukin-6 in mortality from and physiologic response to sepsis. Infect Immun. 2005;73(5):2751ā7.
Lortholary O, Dromer F, Mathoulin-Pelissier S, Fitting C, Improvisi L, Cavaillon JM, Dupont B. French Cryptococcosis Study G: Immune mediators in cerebrospinal fluid during cryptococcosis are influenced by meningeal involvement and human immunodeficiency virus serostatus. J Infect Dis. 2001;183(2):294ā302.
Denham ST, Verma S, Reynolds RC, Worne CL, Daugherty JM, Lane TE, Brown JCS. Regulated release of Cryptococcal Polysaccharide drives virulence and suppresses Immune Cell Infiltration into the Central Nervous System. Infect Immun 2018, 86(3).
Beenhouwer DO, Shapiro S, Feldmesser M, Casadevall A, Scharff MD. Both Th1 and Th2 cytokines affect the ability of monoclonal antibodies to protect mice against Cryptococcus neoformans. Infect Immun. 2001;69(10):6445ā55.
Yoon H, Wake RM, Nakouzi AS, Wang T, Agalliu I, Tiemessen CT, Govender NP, Jarvis JN, Harrison TS, Pirofski LA. Association of Antibody Immunity with Cryptococcal Antigenemia and mortality in a South African cohort with Advanced Human Immunodeficiency Virus Disease. Clin Infect Dis. 2023;76(4):649ā57.
Robertson EJ, Najjuka G, Rolfes MA, Akampurira A, Jain N, Anantharanjit J, von Hohenberg M, Tassieri M, Carlsson A, Meya DB, et al. Cryptococcus neoformans ex vivo capsule size is associated with intracranial pressure and host immune response in HIV-associated cryptococcal meningitis. J Infect Dis. 2014;209(1):74ā82.
Retini C, Vecchiarelli A, Monari C, Tascini C, Bistoni F, Kozel TR. Capsular polysaccharide of Cryptococcus neoformans induces proinflammatory cytokine release by human neutrophils. Infect Immun. 1996;64(8):2897ā903.
Li RK, Mitchell TG. Induction of interleukin-6 mRNA in rat alveolar macrophages by in vitro exposure to both Cryptococcus neoformans and anti-C. Neoformans antiserum. J Med Vet Mycol. 1997;35(5):327ā34.
Tugume L, Ssebambulidde K, Kasibante J, Ellis J, Wake RM, Gakuru J, Lawrence DS, Abassi M, Rajasingham R, Meya DB, et al. Cryptococcal meningitis. Nat Rev Dis Primers. 2023;9(1):62.
Rizzo J, Oliveira DL, Joffe LS, Hu G, Gazos-Lopes F, Fonseca FL, Almeida IC, Frases S, Kronstad JW, Rodrigues ML. Role of the Apt1 protein in polysaccharide secretion by Cryptococcus neoformans. Eukaryot Cell. 2014;13(6):715ā26.
Auge E, Bechmann I, Llor N, Vilaplana J, Krueger M, Pelegri C. Corpora Amylacea in human hippocampal brain tissue are intracellular bodies that exhibit a homogeneous distribution of neo-epitopes. Sci Rep. 2019;9(1):2063.
Martini AC, Helman AM, McCarty KL, Lott IT, Doran E, Schmitt FA, Head E. Distribution of microglial phenotypes as a function of age and Alzheimerās disease neuropathology in the brains of people with Down syndrome. Alzheimers Dement (Amst). 2020;12(1):e12113.
Streit WJ, Braak H, Xue QS, Bechmann I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimerās disease. Acta Neuropathol. 2009;118(4):475ā85.
Barluzzi R, Brozzetti A, Delfino D, Bistoni F, Blasi E. Role of the capsule in microglial cell-Cryptococcus neoformans interaction: impairment of antifungal activity but not of secretory functions. Med Mycol. 1998;36(4):189ā97.
Decken K, Kohler G, Palmer-Lehmann K, Wunderlin A, Mattner F, Magram J, Gately MK, Alber G. Interleukin-12 is essential for a protective Th1 response in mice infected with Cryptococcus neoformans. Infect Immun. 1998;66(10):4994ā5000.
Rincon M, Anguita J, Nakamura T, Fikrig E, Flavell RA. Interleukin (IL)-6 directs the differentiation of IL-4-producing CD4ā+āT cells. J Exp Med. 1997;185(3):461ā9.
Hariri RJ, Chang VA, Barie PS, Wang RS, Sharif SF, Ghajar JB. Traumatic injury induces interleukin-6 production by human astrocytes. Brain Res. 1994;636(1):139ā42.
Huang SH, Wu CH, Chang YC, Kwon-Chung KJ, Brown RJ, Jong A. Cryptococcus neoformans-derived microvesicles enhance the pathogenesis of fungal brain infection. PLoS ONE. 2012;7(11):e48570.
Penkowa M, Giralt M, Lago N, Camats J, Carrasco J, Hernandez J, Molinero A, Campbell IL, Hidalgo J. Astrocyte-targeted expression of IL-6 protects the CNS against a focal brain injury. Exp Neurol. 2003;181(2):130ā48.
Lee SC, Dickson DW, Brosnan CF, Casadevall A. Human astrocytes inhibit Cryptococcus neoformans growth by a nitric oxide-mediated mechanism. J Exp Med. 1994;180(1):365ā9.
Trajkovic V, Stepanovic S, Samardzic T, Jankovic V, Badovinac V, Mostarica Stojkovic M. Cryptococcus neoformans neutralizes macrophage and astrocyte derived nitric oxide without interfering with inducible nitric oxide synthase induction or catalytic activity - possible involvement of nitric oxide consumption. Scand J Immunol. 2000;51(4):384ā91.
Klein MA, Moller JC, Jones LL, Bluethmann H, Kreutzberg GW, Raivich G. Impaired neuroglial activation in interleukin-6 deficient mice. Glia. 1997;19(3):227ā33.
Giles SS, Zaas AK, Reidy MF, Perfect JR, Wright JR. Cryptococcus neoformans is resistant to surfactant protein A mediated host defense mechanisms. PLoS ONE. 2007;2(12):e1370.
Coelho C, Camacho E, Salas A, Alanio A, Casadevall A. Intranasal Inoculation of Cryptococcus neoformans in Mice Produces Nasal Infection with Rapid Brain Dissemination. mSphere 2019, 4(4).
Chen M, Liao WQ, Wu SX, Yao ZR, Pan WH, Liao Y. Taxonomic analysis of cryptococcus species complex strain S8012 revealed Cryptococcus gattii with high heterogeneity on the genetics. Chin Med J (Engl). 2011;124(13):2051ā6.
Kidd SE, Hagen F, Tscharke RL, Huynh M, Bartlett KH, Fyfe M, Macdougall L, Boekhout T, Kwon-Chung KJ, Meyer W. A rare genotype of Cryptococcus gattii caused the cryptococcosis outbreak on Vancouver Island (British Columbia, Canada). Proc Natl Acad Sci U S A. 2004;101(49):17258ā63.
Ibe C, Okoye CA, Nweze E, Otu A. Cryptococcosis in Africa: what the data tell us. Med Mycol 2023, 61(6).
Huang M, Pang X, Karalis K, Theoharides TC. Stress-induced interleukin-6 release in mice is mast cell-dependent and more pronounced in apolipoprotein E knockout mice. Cardiovasc Res. 2003;59(1):241ā9.
Dufaud C, Rivera J, Rohatgi S, Pirofski LA. Naive B cells reduce fungal dissemination in Cryptococcus neoformans infected Rag1(-/-) mice. Virulence. 2018;9(1):173ā84.
Meyerholz DK, Sieren JC, Beck AP, Flaherty HA. Approaches to evaluate lung inflammation in Translational Research. Vet Pathol. 2018;55(1):42ā52.
Aslanyan L, Ekhar VV, DeLeon-Rodriguez CM, Martinez LR. Capsular specific IgM enhances complement-mediated phagocytosis and killing of Cryptococcus neoformans by methamphetamine-treated J774.16 macrophage-like cells. Int Immunopharmacol. 2017;49:77ā84.
Aslanyan L, Lee HH, Ekhar VV, Ramos RL, Martinez LR. Methamphetamine impairs IgG1-Mediated phagocytosis and killing of Cryptococcus neoformans by J774.16 macrophage- and NR-9640 Microglia-Like cells. Infect Immun 2019, 87(2).
Acknowledgements
The microglial cell line derived from wild-type mice, NR-9460, was obtained through BEI Resources (NIAID, NIH).
Funding
M. R-G, M. E. M., S. D., M. F. H., and L.R.M. were supported by the National Institute of Allergy and Infectious Diseases (NIAID award # R01AI145559) of the US National Institutes of Health (NIH). M. E. M was supported by the UFCDās Comprehensive Training Program in Oral Biology (NIH NIDCR Award # T90DE021990/R90DE022530). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Author information
Authors and Affiliations
Contributions
M. R.-G. designed and performed experiments, collected and analyzed data, prepared the figures, and wrote the manuscript. M. E. M. performed experiments and wrote the manuscript. M. F. H. examined and analyzed the histological images.C. C.-N. designed experiments and collected and analyzed data.L. R. M. designed experiments, analyzed data, prepared the figures, wrote the manuscript, and secured funding.
Corresponding author
Ethics declarations
Competing interests
L.R.M. reports a patent (issued and pending) assigned to the University of Florida. All other authors report no potential conflicts.
Additional information
Publisherās note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
12974_2024_3237_MOESM1_ESM.tif
Supplementary Material 1: Wild-type, IL-6ā/ā, IL-6ā/ā + rIL-6 mice systemically infected with Cn showed no difference in pulmonary pathology development. Mice were infected IV with 105 cryptococci and euthanized at (A) 3- and (B) 7-dpi. Representative images of lung tissue sections stained with periodic acid-Schiff. Black, yellow, blue, and green arrows indicate normal, atelectasis, hyperplasia, and cryptococcoma formation, respectively. Red arrowheads denote bronchus-associated lymphoid tissue. Top, middle, and bottom panels indicate 4, 10, and 20X magnification, respectively. Scale bars: 50 Ī¼m.
12974_2024_3237_MOESM2_ESM.tif
Supplementary Material 2: Wild-type mice infected with Cn show increased IL-6 levels in the brain compared to uninfected mice. The supernatants from uninfected and H99-infected Wild-type (C57BL/6) brains at 3- and 7-dpi were processed and analyzed for IL-6 levels by ELISA. Bars represent the mean values and error bars indicate SDs. Each circle represents supernatant from an individual brain (nĀ =Ā 3 supernatants per group). Significance (****, PĀ <Ā 0.0001; ***, PĀ <Ā 0.001; **, PĀ <Ā 0.01) was calculated by one-way ANOVA and adjusted using Tukeyās post-hoc analysis. ns denotes comparisons which are not statistically significant. Cytokine quantification was performed twice with similar results obtained.
12974_2024_3237_MOESM3_ESM.tif
Supplementary Material 3: siRNA treatment does not affect NR-9460 microglia-like cell viability. (A) Representative flow cytometry dot plots for untreated and siRNA IL-6- or siRNA negative control-treated NR-9460 cells are shown. Cells were stained with 7-AAD for viability after a 24 h siRNA treatment at 37Ā°C and 5% CO2. Each plot was generated after ā„Ā 10,000 events were analyzed. (B) The percentage of microglia-like cell viability was determined. Each symbol represents an independent replicate (nĀ =Ā 4). Bars and error bars denote means and SDs, respectively. Significance (*, PĀ <Ā 0.05) was calculated by one-way ANOVA and adjusted using Tukeyās post-hoc analysis. ns denotes comparisons which are not statistically significant.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the articleās Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the articleās Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Reguera-Gomez, M., Munzen, M.E., Hamed, M.F. et al. IL-6 deficiency accelerates cerebral cryptococcosis and alters glial cell responses. J Neuroinflammation 21, 242 (2024). https://doi.org/10.1186/s12974-024-03237-x
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s12974-024-03237-x