Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Latest Articles
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • Topics
    • Applied and Environmental Science
    • Clinical Science and Epidemiology
    • Ecological and Evolutionary Science
    • Host-Microbe Biology
    • Molecular Biology and Physiology
    • Therapeutics and Prevention
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About mBio
    • Editor in Chief
    • Board of Editors
    • AAM Fellows
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
mBio
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Latest Articles
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • Topics
    • Applied and Environmental Science
    • Clinical Science and Epidemiology
    • Ecological and Evolutionary Science
    • Host-Microbe Biology
    • Molecular Biology and Physiology
    • Therapeutics and Prevention
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About mBio
    • Editor in Chief
    • Board of Editors
    • AAM Fellows
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
Research Article | Host-Microbe Biology

Serum Amyloid P Component Binds Fungal Surface Amyloid and Decreases Human Macrophage Phagocytosis and Secretion of Inflammatory Cytokines

Nicole E. Behrens, Peter N. Lipke, Darrell Pilling, Richard H. Gomer, Stephen A. Klotz
Michael Lorenz, Editor
Nicole E. Behrens
aDepartment of Immunobiology, University of Arizona, Tucson, Arizona, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Peter N. Lipke
bDepartment of Biology, Brooklyn College, Brooklyn, New York, USA
cGraduate Center of the City University of New York, New York, New York, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Darrell Pilling
dDepartment of Biology, Texas A&M University, College Station, Texas, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Richard H. Gomer
dDepartment of Biology, Texas A&M University, College Station, Texas, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stephen A. Klotz
eDepartment of Medicine, University of Arizona, Tucson, Arizona, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael Lorenz
University of Texas Health Science Center
Roles: Editor
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joel Buxbaum
Scripps Institute
Roles: Solicited external reviewer
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marinos Kallikourdis
Humanitas University
Roles: Solicited external reviewer
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/mBio.00218-19
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

In patients with invasive fungal diseases, there is often little cellular inflammatory response. We tested the idea that binding of the human constitutive plasma protein serum amyloid P component (SAP) (also called PTX2) to Candida albicans dampens the innate immune response to this fungus. Many pathogenic fungi have cell surface amyloid-like structures important for adhesion and biofilm formation. Human SAP bound to fungi that expressed functional cell surface amyloid, but SAP had minimal binding to fungi with reduced expression of cell surface amyloid. In the absence of SAP, phagocytosis of fungi by human macrophages was potentiated by expression of amyloid on the fungi. SAP binding to fungi inhibited their phagocytosis by macrophages. Macrophages pretreated with SAP displayed reduced fungal phagocytosis, reduced secretion of inflammatory cytokines (IFN-γ, IL-6, and TNF-α), and increased secretion of the anti-inflammatory cytokine IL-10. SAP bound to fungi or added to the medium upregulated the expression of the anti-inflammatory receptor CD206 on macrophages. These findings suggest that SAP bound to amyloid-like structures on fungal cells dampens the host cellular immune response in fungal diseases such as invasive candidiasis.

IMPORTANCE Macrophages are a key part of our innate immune system and are responsible for recognizing invading microbes, ingesting them, and sending appropriate signals to other immune cells. We have found that human macrophages can recognize invading yeast pathogens that have a specific molecular pattern of proteins on their surfaces: these proteins have structures similar to the structures of amyloid aggregates in neurodegenerative diseases like Alzheimer’s disease. However, this surface pattern also causes the fungi to bind a serum protein called serum amyloid P component (SAP). In turn, the SAP-coated yeasts are poorly recognized and seldom ingested by the macrophages, and the macrophages have a more tolerant and less inflammatory response in the presence of SAP. Therefore, we find that surface structures on the yeast can alter how the macrophages react to invading microbes.

INTRODUCTION

In humans, the pentraxin serum amyloid P component (SAP) is constitutively present in serum (typically ∼30 µg/ml) (1, 2). SAP binds to amyloid fibrils derived from precursors, including serum amyloid A, immunoglobulin light chain, and apolipoprotein A1, and also binds to pathogenic viruses (3) and bacteria (4, 5). The binding of SAP to microorganisms may be an important factor in determining the outcome of some disseminated infectious diseases. For example, one report demonstrated that bacteria to which SAP bound were not opsonized or phagocytosed efficiently (5). In mouse models of disseminated infection with Escherichia coli, Streptococcus pyogenes, or Neisseria meningitidis (all of which bound SAP), the bacteria were lethal when injected intravenously. SAP knockout mice survived infections with similar numbers of the same bacteria, but when SAP knockout mice were injected with microbes and human SAP, the mice succumbed to disseminated infection (5). Thus, the presence of SAP can increase virulence in bacterial infection models.

SAP avidly binds to amyloid and is a prominent and invariant constituent of all extracellular amyloid deposits (6). We have proposed that SAP also binds to fungi because of functional amyloid on fungal cell surfaces (7). Candida albicans cell surface adhesins form amyloid-like nanodomains (8). These amyloid nanodomains form in response to shear stress and are essential for cell-cell aggregation and biofilm formation (9). The amyloid interactions are functional, in the sense that amyloid-forming ability is an important part of adhesin activity and is evolutionarily conserved in fungal adhesins and some bacterial adhesins (10). We previously observed SAP on fungi in invasive Candida in human tissue and found that SAP binding to the fungi, in part, required the presence of functional amyloid on the surfaces of fungal cells (11). We also observed SAP on fungal surfaces in deep-seated fungal diseases, including aspergillosis, coccidioidomycosis, and zygomycosis (12).

Polymorphonuclear leukocytes (PMNs) and macrophages are sparse in anatomic sites of invasive candidiasis even in cases with normal or elevated white blood cell counts (11, 13). Marked reductions of host immune cells were also seen in deep-seated disease with Aspergillus, zygomycetes, and Coccidioides (12). Other investigators have also noted the profound absence of host immune cells in the vicinity of invasive fungi including aspergillosis and mucormycosis, again in spite of normal or elevated peripheral white blood cell counts (14–16). Thus, leucopenia alone cannot explain the lack of cellular infiltrates. Pentraxin 3 and C reactive protein (CRP), two pentraxins generally acknowledged to be proinflammatory, were not detected on Candida species invading gastrointestinal tissue (13). This apparent lack of an innate immune response is reminiscent of the histology of extracellular amyloid deposits, with their absence of any detectable cellular response even as the amyloid destroys organs (17). A commonality between these conditions is SAP binding to amyloid in extracellular deposits or amyloid on fungal surfaces.

Here we report that SAP binding to C. albicans leads to decreased phagocytosis by human macrophages and to a less aggressive host innate cellular response. These changes may be a molecular explanation for the histological findings of minimal inflammation in invasive fungal diseases.

RESULTS

SAP binds to yeast cells in proportion to the amount of functional amyloid expressed on the fungal surface.To elucidate SAP binding to C. albicans, yeast cells were exposed to SAP from normal human male AB serum by incubating yeasts in serum for 1 h (the most physiological method of providing SAP, avoiding the problem of SAP aggregation [18]). Figure 1A shows a flow cytometric profile demonstrating that most C. albicans cells bound SAP and there was minimal background fluorescence. We then compared SAP binding to C. albicans to SAP binding to nonpathogenic laboratory strains of Saccharomyces cerevisiae. S. cerevisiae W303-1B transformed with an empty vector (pJL1) (hereinafter, S. cerevisiae-EV) expresses low levels of endogenous amyloid, as does a transformant that expresses the nonamyloidogenic Als5pV326N form of the C. albicans adhesin Als5p (S. cerevisiae-Als5pV326N) (19). Heterologous expression of the amyloidogenic adhesin Als5pWT in S. cerevisiae (S. cerevisiae-Als5pWT) causes yeast cells to adhere and form biofilms in a manner similar to that of C. albicans (20, 21). Figure 1B shows that a median of 92% of C. albicans bound SAP. By comparison, only 18% of S. cerevisiae-EV cells bound SAP; S. cerevisiae-Als5pV326N, 19% of cells; the amyloidogenic S. cerevisiae-Als5pWT, 40% of cells. Thus, SAP bound to yeast cells in the order of their expression of cell surface amyloid: C. albicans > S. cerevisiae-Als5pWT ≫ S. cerevisiae-Als5pV326N or S. cerevisiae-EV.

FIG 1
  • Open in new tab
  • Download powerpoint
FIG 1

Binding and phagocytosis of yeasts by human macrophages. (A) SAP binding to C. albicans yeast cells determined by SAP antibody labeling and flow cytometry. (B) SAP binding to yeast cells of C. albicans, S. cerevisiae-EV, the amyloidogenic S. cerevisiae-Als5pWT, and the nonamyloidogenic, S. cerevisiae-Als5pV326N. SAP was offered to yeasts in normal human serum. Each symbol is the average of one of seven independent experiments. (C, left) Binding of fungi to macrophages upon the addition of C. albicans, S. cerevisiae-EV, S. cerevisiae-Als5pWT, or S. cerevisiae-Als5pV326N. (Right) Phagocytosis of the same strains of yeast cells by macrophages. Each symbol is the average from three independent experiments. (D) Effect of SAP on macrophage binding and phagocytosis of C. albicans. (Top left) Binding of yeast cells to control macrophages or macrophages pretreated with SAP. (Top right) Phagocytosis of C. albicans by control macrophages or macrophages pretreated with SAP (30 µg/ml). (Bottom left) Binding of yeast cells in serum-free medium and yeast cells offered SAP in normal human serum to macrophages. (Bottom right) Phagocytosis by macrophages of yeast cells in serum-free medium and yeast cells offered SAP in normal human serum (results of three separate experiments each with 3,200 macrophages counted/variable). Note that the y axis is 10-fold higher for phagocytosis than the binding graphs. Statistical significance was determined by Mann-Whitney U test and indicated as follows: ns, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. Medians are plotted to show central tendency. Bars represent 95% confidence intervals.

SAP binding to C. albicans is inhibited by the calcium chelator EDTA.SAP binding to amyloid fibers is calcium dependent, and bound SAP is dissociated from amyloid fibers with EDTA (18). To determine whether SAP binding to C. albicans is also sensitive to EDTA, we added EDTA to mixtures of serum and C. albicans. EDTA significantly inhibited SAP binding to the cells (Table 1) (see Fig. S1A in the supplemental material). These data suggest that SAP binding to C. albicans is at least partly dependent on calcium.

View this table:
  • View inline
  • View popup
TABLE 1

Inhibition or augmentation of yeast binding of SAP and phagocytosis by human macrophages in the presence or absence of various additivesa

FIG S1

Phagocytosis of C. albicans yeasts by human macrophages under different treatment conditions. Download FIG S1, DOCX file, 0.2 MB.
Copyright © 2019 Behrens et al.

This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.

Phagocytosis of fungi is dependent on expression of fungal functional amyloid.We investigated the phagocytosis of yeasts by human macrophages and found that macrophages bound approximately twice as many C. albicans cells as S. cerevisiae cells. The three S. cerevisiae strains bound in similar numbers (Fig. 1C, left). However, the macrophages ingested significantly different numbers of the yeasts (Fig. 1C, right). Macrophages phagocytosed a median of 79 C. albicans yeast cells/50 macrophages and 57 cells/50 macrophages for the amyloid-bearing strain S. cerevisiae-Als5pWT. In contrast, few non-amyloid-bearing yeasts were ingested: S. cerevisiae-EV, 4 cells/50 macrophages and S. cerevisiae-Als5pV326N, 17 cells/50 macrophages. These data suggest that the presence of yeast cell surface amyloid increases the phagocytosis of yeast cells.

SAP treatment of macrophages inhibits macrophage phagocytosis of yeast cells.Having shown that SAP binding to yeast cells and macrophage phagocytosis of yeast cells are both increased by the presence of yeast surface amyloids, we investigated the effect of SAP on macrophage phagocytosis of yeast cells. Human macrophages were incubated with or without SAP for 60 min in 96-well plates, the wells were washed with serum-free medium, and then yeasts were added for 30 min in serum-free medium. The SAP pretreatment led to an ∼3-fold increase, from ∼4 to ∼11 C. albicans cells bound per 50 macrophages (Fig. 1D, top left), but a 86% reduction in macrophage phagocytosis, from a median of 124 yeast cells phagocytosed/50 macrophages in control wells (no SAP) to 17 yeast cells/50 macrophages in wells with SAP (Fig. 1D, top right). In contrast, pretreating macrophages with the related pentraxin, C-reactive protein (CRP) (50 µg/ml) enhanced yeast phagocytosis by 16% (Fig. S1B). CRP bound to negligible numbers of yeast cells on flow cytometry (7%). SAP from Millipore contained low concentrations of azide and EDTA; both of the additives at various concentrations were incubated with macrophages to determine whether these additives affected phagocytosis, and they did not (Fig. S1C). Furthermore, phagocytosis results were not affected using unaltered SAP from Millipore or SAP that had been desalted (removing any trace of azide, EDTA, and NaCl) (Fig. S1B and C). Together these data support the hypothesis that SAP treatment of the macrophages inhibits phagocytosis of C. albicans.

Only 10% to 40% of macrophages phagocytosed yeast (Fig. 2A). The morphology of SAP-treated macrophages was noticeably different from that of control macrophages: the former were smaller and more compact with rounded cell contours rather than pseudopodal extensions. This may be an attribute of the immunological quiescence of the cells due to SAP treatment (22, 23). A time course showed that SAP treatment of the macrophages was rapid, with >90% inhibition of phagocytosis after 60 min of incubation with exogenously added SAP prior to the addition of yeasts (Fig. 2C). Because mannose is an inhibitor of SAP binding to macrophages (24), a major saccharide found in the cell wall of fungi, and the mannose receptor found on macrophages is important in yeast phagocytosis, we tested the effect of D-mannose on the interaction of yeasts and macrophages. Phagocytosis of yeasts was inhibited by 78% in the presence of 100 mM mannose (Table 1), implying involvement of the macrophage mannose receptor.

FIG 2
  • Open in new tab
  • Download powerpoint
FIG 2

Phagocytosis of yeast. (A) Macrophages in serum-free medium (pink cytoplasm, ∼40 μm in diameter) and untreated C. albicans yeast cells (blue ovals, ∼2 to 3 μm existing in singlets and doublets and multiples of the same). Many yeasts are phagocytosed by macrophages. Bar, 50 µm. (Upper right) Close-up of macrophage that has phagocytosed 15 yeast cells. Bar, 50 µm. (B) Photomicrograph of macrophages pretreated with SAP. Note the round conformation of the macrophages compared to those in panel A above. Bar, 50 µm. (C) Phagocytosis of yeasts by macrophages preincubated in serum-free medium in the presence or absence of SAP (30 µg/ml). Results represent yeasts phagocytosed by 400 macrophages per time point. Results at zero and 15 min are not significantly different. The time points of 30, 60, and 120 min are significantly different with P values of <0.05, <0.01, and <0.01, respectively. Medians are depicted to show central tendency. Bars represent 95% confidence limits, and significance was determined by the Mann-Whitney U test.

Serum or SAP treatment of yeast inhibits macrophage phagocytosis of yeast.To determine whether incubation of yeast with serum affects macrophage phagocytosis of yeast, Candida albicans yeasts were incubated in normal human serum for 1 h, washed, and then incubated with human macrophages. Compared with pretreatment of the macrophages with SAP, serum treatment of the yeasts did not significantly increase binding of yeasts to the macrophages (Fig. 1D, bottom left). Nevertheless, serum treatment of the yeasts decreased macrophage phagocytosis of yeast by 82% (Fig. 1D, bottom right). This result is not significantly different from the 86% inhibition of phagocytosis following pretreatment of macrophages with SAP. The same phenomenon occurred with macrophages cultured on chamber slides (Table 1). In addition, macrophage phagocytosis of yeast decreased by 62% when the yeast cells were presoaked in purified SAP for 1 h in TBS-C (Table 1 and Fig. S1D). These results indicate that pretreatment of yeast or macrophages with SAP profoundly inhibits macrophage phagocytosis of yeast. The results were consistent for SAP added as a constituent of serum or as purified SAP in buffer.

The combination of SAP and Candida increases expression of two macrophage M2 markers.Macrophages change markers and physiology when exposed to different external signals (25). We assessed the effects of SAP and yeasts on macrophage markers associated with polarization: ICAM-1 (CD54), CD206, CD209, and the transcription factors IRF4 and IRF5. All macrophages showed expression of CD45, demonstrating their origin from the bone marrow (Fig. 3A). There were no significant effects of Candida, SAP, or serum on the percentage of cells expressing the two M1 markers ICAM-1 and IRF5 (Fig. 3A). Examining the correlation between yeast phagocytosis and marker expression, we observed that all macrophages that phagocytosed yeasts expressed ICAM-1. SAP and Candida plus SAP increased the percentage of cells expressing the M2 marker CD206, and Candida plus SAP and serum-soaked Candida increased the percentage of cells expressing IRF4 (Fig. 3B). SAP decreased the percentage of cells expressing the M2 marker CD209 (Fig. 3B). Together, these results indicate that SAP and Candida can affect some marker expression in macrophages.

FIG 3
  • Open in new tab
  • Download powerpoint
FIG 3

Expression of markers by macrophages. (A and B) Macrophages were incubated in serum-free medium (control) or in serum-free medium plus the following: C. albicans alone; SAP alone; SAP and C. albicans; or C. albicans yeast cells offered SAP in normal human serum. Results show the percentage of macrophages staining for markers following different treatments. A bar within a colored box is the median value, and the box represents the 95% confidence intervals of values from four separate donors. Values that are significantly different (P < 0.05) by Mann-Whitney U test are indicated by a bar and asterisk.

Compared to Candida alone, SAP and Candida decrease accumulation of the proinflammatory cytokine IFN-γ and increase accumulation of the anti-inflammatory cytokine IL-10.Macrophage signaling through secreted cytokines is a key event in innate and acquired immunity. Therefore, we assessed extracellular cytokine accumulation in the presence of C. albicans yeasts and/or SAP. Macrophage extracellular accumulation of IL-6, IFN-γ, TNF-α, and IL-10 was increased by either yeasts or SAP (Fig. 4). However, compared to SAP alone or yeast alone, combining SAP and yeast decreased the accumulation of the inflammatory cytokine IFN-γ and increased the anti-inflammatory cytokine IL-10 (Fig. 4). Extracellular IL-17A accumulation was increased when yeasts were added to the cell culture, and SAP did not significantly affect the accumulation of this cytokine (Table 2).

FIG 4
  • Open in new tab
  • Download powerpoint
FIG 4

Cytokine secretion by macrophages after various treatments. The treatments were : macrophages in serum-free medium (Mac. Only), addition of C. albicans to macrophages (Post Can.), addition of SAP only (post SAP Pre Can.), and pretreatment of macrophages with SAP (30 µg/ml) followed by the addition of C. albicans (Post SAP. Post Can.). Medians are plotted to show central tendencies with 95% confidence intervals. Statistical significance was determined by Mann-Whitney U test and indicated as follows: ns, not significant; *, P < 0.05; **, P < 0.01.

View this table:
  • View inline
  • View popup
TABLE 2

IL-17A secretion by macrophages in response to different treatmentsa

SAP does not inhibit PMN phagocytosis of yeasts.Because PMNs are important in cellular innate immunity, we examined what effect SAP has on PMN phagocytosis of C. albicans. Human PMNs were pretreated with SAP (30 µg/ml) for 60 min, and C. albicans yeasts were then added in serum-free medium. After 30 min of coincubation, there was no significant effect of SAP on PMN phagocytosis of yeast, and this effect was seen for both male and female donors (Fig. 5).

FIG 5
  • Open in new tab
  • Download powerpoint
FIG 5

Phagocytosis of C. albicans yeasts by human polymorphonuclear leukocytes (PMNs). Controls are PMNs offered yeasts in serum-free medium; SAP treated are PMNs pretreated with SAP (30 µg/ml), washed, and offered yeasts in serum-free medium. Cells were from three females (F-1 to F-3) and two males (M-1 and M-2). Medians are plotted to show central tendency with 95% confidence levels. There was no significant difference with the Mann-Whitney U test between controls and SAP-pretreated PMNs with respect to phagocytosis.

DISCUSSION

Results reported here may explain the highly attenuated inflammatory response in infected tissue, a paradoxical feature of invasive fungal disease in humans (11, 13–16). In several invasive fungal diseases, fungal cells in deep infections display surface amyloid-like structures and are coated by the amyloid-binding SAP (11–13). We now report that a consequence of SAP binding to fungi is the down-modulation of the innate cellular immune response.

Human macrophages recognize fungal surface amyloid-like structures.Macrophages bind yeasts through lectin receptors, including the mannose receptor (MR) (CD206) and DC-SIGN or CD209 (26, 27) to facilitate phagocytosis even when bacteria or yeasts are unopsinized (27, 28). Our data reflect the involvement of CD206, because phagocytosis was significantly inhibited by the addition of D-mannose. Under the conditions of our experiments, phagocytosis of yeasts was clearly related to the level of surface amyloids present on the fungi, but how much occurs through CD206 or DC-SIGN is unknown. Macrophages were largely indifferent to our laboratory strain of S. cerevisiae which expresses very low background levels of the amyloidogenic proteins Flo1 and Flo11 (29), and there was minimal phagocytosis of yeast expressing the nonamyloidogenic Als5pV326N (19). In contrast, yeast cells expressing the wild-type amyloidogenic sequence were phagocytosed at high levels. C. albicans yeast cells were phagocytosed to an even higher degree, and these cells possess a higher level of surface amyloid and a large number of potentially amyloidogenic adhesins (9, 10, 30). Therefore, formation of amyloid-like nanodomains could activate phagocytosis by clustering CD206 into high-avidity patches on the cell surface, as we have shown for activation of fungal adhesins (9). Bacterial functional amyloids have frequently been implicated as virulence factors in many diseases including involvement with innate immune cells (31). One example is the amyloid pilus of Mycobacterium tuberculosis that facilitates invasion of macrophages (32), and SAP markedly reduces M. tuberculosis phagocytosis (33). The phagocytosis of M. tuberculosis is similar to that of C. albicans, i.e., both are engulfed within a phagolysosome, the difference is that the bacteria persist in macrophages, whereas C. albicans is killed (34), or the fungus kills the macrophage after shifting to hyphal morphology (35). Phagocytosis of M. tuberculosis or Mycobacterium smegmatis in the presence of SAP also skews the macrophages toward the “tolerant” M2c cell type, and this skew can be reduced in the presence of small molecules that block SAP binding (36). Thus, in both microbes (M. tuberculosis and C. albicans), SAP may bind to amyloid-like structures and lead to increased microbe survival and propagation.

SAP binding to fungi is dependent upon fungal expression of functional amyloid and is antiphagocytic.The importance of SAP binding to bacteria and the implications this phenomenon might have on host defenses were reported years ago (5), but they have not been pursued systematically. It was proposed that binding of SAP to beads is opsonic, but SAP binding to bacteria protects the bacteria from degradation and is antiopsonic and anti-immunogenic (5, 37). Binding of SAP to C. albicans may give rise to similar antiphagocytic and anti-inflammatory effects. Wild-type C. albicans expresses a number of cell surface functional amyloids, the best studied is Als5p (19), a member of a family of amyloidogenic adhesive proteins variably expressed on the cell surface (7). In invasive candidiasis, SAP binds to functional amyloid nanodomains on the surface of the fungi. The consequences of this binding include inhibition of macrophage phagocytosis and reduction in secretion of inflammatory cytokines. Thus, human SAP, a soluble pentraxin that binds avidly to pathological human amyloids (38), also binds to C. albicans functional amyloids.

The presence of functional amyloids explains the binding of SAP to S. cerevisiae-Als5p compared to S. cerevisiae-EV and S. cerevisiae-Als5pV326N, which has little surface amyloid. The SAP binding capacities of the yeasts were C. albicans > S. cerevisiae-Als5pWT ≫ S. cerevisiae-Als5pV326N > S. cerevisiae-EV, an order consistent with the ability of yeasts to form amyloid-like surface nanodomains (9, 10, 39). The low levels of SAP binding could be due to residual surface amyloids (39) or exposure of phosphoethanolamine and/or phosphocholine in the yeast cell wall, as they are ligands of SAP (40–42). It is possible that SAP binding to microbial amyloid is common: SAP binding to surface proteins of Neisseria meningitidis was investigated, and two of the three identified SAP-binding proteins, NMB0667 and NMB201 (43), have multiple regions with high amyloid potential, like many fungal adhesins (data not shown).

SAP coating of fungi markedly inhibited phagocytosis, with a corresponding increase in C. albicans bound but not engulfed. The results were similar whether the yeasts or macrophages were treated with purified SAP or SAP was offered to yeasts in normal serum, and the effects of SAP on macrophage phagocytosis were detected within minutes. This SAP binding may result from fungal exploitation of SAP’s role as an immunomodulatory molecule, rather than as a human adaptation to infection. Fungal surface amyloids have evolved as components of biofilms, and SAP binding may be a fortuitous consequence (8, 9, 11, 23, 38).

SAP is anti-inflammatory.The consequences of SAP binding included reduction in accumulation of proinflammatory cytokines and an increase in accumulation of anti-inflammatory IL-10. IL-10 secretion by macrophages increased in response to SAP treatment of the macrophages or the yeast cells. These results confirm well-established anti-inflammatory properties of SAP, which has been previously shown to reduce neutrophil adhesion (44, 45), to inhibit maturation of monocytes to fibrocytes in tissue (44), and to coat cell debris which is scavenged by macrophages (37). Accordingly, SAP also directs macrophage differentiation toward resolving macrophage class M2c (22). The anti-inflammatory effect is dependent upon the production of IL-10 which follows SAP binding to DC-SIGN (46).

Our incubation of SAP with macrophages in fluid phase or bound to C. albicans yeasts led to changes in macrophage surface antigen expression. Markers of the M1 phenotype (ICAM-1, IFR5) were unchanged, whereas IRF4 and the M2 marker phenotype CD206 increased. Detection of CD209 (DC-SIGN) decreased with exposure to SAP. CD209 is a macrophage receptor that may account for up 30% of C. albicans phagocytosis (28), and SAP binding to DC-SIGN leads to increased IL-10 secretion that potentiates an anti-inflammatory response (46). Detection of DC-SIGN with antibodies was lower on the macrophages incubated with SAP, either due to downregulation of the receptor, or perhaps the antibody binding site was masked by the binding of SAP to the receptor. These results are in conformity with the anti-inflammatory properties of SAP (23) and along with the changes in cytokine expression due to SAP, may help to downregulate the response of the innate immune system.

However, CD206 (MR receptor) was moderately upregulated in the presence of SAP. Candida binding to MR (CD206) stimulates macrophages to secrete the proinflammatory cytokine IL-17 (47), a feature we observed. The amount of IL-17 secreted was not altered by incubating the macrophages with SAP, but it was the only proinflammatory cytokine observed to increase in the presence of SAP and yeasts. Thus, we find that for invasive candidiasis, the presence of SAP on macrophages or yeasts, shifted the macrophage response toward an anti-inflammatory outcome.

PMNs and SAP.We did not find SAP-dependent differences in neutrophil phagocytosis of C. albicans. SAP reduces neutrophil adhesion and spreading (23), and this may have led to the very small numbers of yeasts phagocytosed.

Conclusion.SAP binds to C. albicans fungal cell surface functional amyloid, a structure recognized by macrophages. SAP bound to fungal surfaces is antiphagocytic and anti-inflammatory, reducing phagocytosis of fungi and increasing IL-10 secretion (Fig. 6). Although the question remains open whether SAP binding to fungi helps the host or the pathogen (5, 48), it appears from our work that SAP-coated yeasts are either masked from the innate immune system or, upon contact with host cells, dampen the immune response.

FIG 6
  • Open in new tab
  • Download powerpoint
FIG 6

Figurative rendition of important findings of this investigation involving human macrophage-SAP-Candida albicans yeast interactions. (Left) The macrophage is a control macrophage (cultured in serum-free medium) and C. albicans yeasts added to it in serum-free medium. The cell secretes numerous cytokines and phagocytoses yeasts in great numbers. (Top right) The macrophage is cultured like the control macrophage, but the C. albicans yeasts are soaked in normal human serum or in purified SAP for an hour before addition to the macrophage, allowing SAP to bind to the fungal functional amyloid. Phagocytosis is reduced >80%. (Bottom right) The macrophage is cultured identically to the control macrophage except that it is incubated with SAP (30 µg/ml) for 1 h before C. albicans yeasts are added. Like the macrophage above it, there is a >80% reduction in phagocytosis of yeast cells, but only IL-10 and IL-17A are secreted in increased amounts. After SAP treatment, the macrophages were more rounded in their overall shape, a reflection of their quiescence.

MATERIALS AND METHODS

Macrophages.Human macrophages were isolated from whole blood using the SepMate-50 system (Stemcell Technologies, Seattle, WA) and following a University of Arizona IRB-approved protocol with written consent from the donors. After isolation, the cells were cryopreserved gradually with a controlled rate freezer. For experimental use, cryopreserved peripheral blood mononuclear cells (PBMCs) from normal (healthy) adult controls were placed in complete RPMI (RPMI, 10% fetal bovine serum, penicillin/streptomycin, and DNase [3 ng/ml]) and thawed to 37°C. Cells were collected by centrifugation at 500 × g for 6 min, resuspended in X-VIVO-15 (Lonza, Walkersville, MD), a serum-free medium, plated into 96-well plates (Fisher Scientific) (105 cells in 100 µl medium/well), and placed in an incubator overnight at 37°C with 5% CO2. The following day PBMCs were washed with serum-free medium and resuspended in fresh medium, human MCSF (Sigma-Aldrich, St. Louis, MO) was added to each well (1 ng/ml), and cells were incubated for 5 days at 37°C with 5% CO2. Experiments were also carried out in four-well slide chambers (Bio Express, Visalia, CA) with the same medium and additives and 200 µl/well. Slide chambers allowed for better photomicroscopy of the macrophage-fungus interactions.

Fungi.Two Candida albicans strains were used in this study, one a clinical isolate (Banner University Medical Center, Tucson, AZ) and the other, a quality control strain from the Clinical and Laboratory Standards Institute (Wayne, PA). There was no difference in results with the two strains. The two strains were maintained on YPD agar (RPI, Mount Pleasant, IL). For experiments, a loopful of fungi was added to 5 ml YPD broth (Life Technologies Corp., Carlsbad, CA) and incubated overnight at 26°C with shaking. After 24 h, the yeasts were collected by centrifugation at 1,200 × g for 5 min, resuspended, and washed thoroughly in Tris-buffered saline with 2 mM Ca2+ (TBS-C). This was repeated three times, and cells were then resuspended to 109 cells/ml in TBS-C and used as described below. Saccharomyces cerevisiae strains Als5p, V326N, and EV (background W303-1B) expressing variations of the C. albicans Als5p protein (19) were under a galactose promoter; therefore, the strains were grown in CSM-Trp galactose broth plus adenine (Thermo Fisher, Waltham, MA). Yeast strains were maintained on agar with the same medium. S. cerevisiae strains were cultured in CSM-Trp galactose broth plus adenine at 26°C for 48 h, and washed with TBS-C as described above.

Macrophage-yeast interactions.Wells containing 5-day-old macrophages in chamber slides or 96-well plates were washed with serum-free medium, and 100 µl of fresh serum-free medium and 1 µl containing 106 previously washed yeasts suspended in TBS-C were added to each well. For other experiments, alongside the addition of C. albicans alone to wells or chambers, C. albicans was also incubated in 100% normal human male AB serum (Innovative Research, Novi, MI) or SAP (Millipore, Temecula, CA) at 30 µg/ml for 1 h at 4°C, washed with TBS-C, resuspended in serum-free medium, and added to wells or chambers. The cocultures were incubated at 37°C on a rocking shaker for 30 min. The yeast/macrophage ratio varied from 100 to 10:1. A 30-min incubation of yeasts with macrophages was employed to minimize the number of yeasts forming germ tubes. Following incubation of yeasts with macrophages, the wells were washed gently three times with TBS-C, and the wells or slides were stained with Wright-Giemsa. Stained wells and slides were observed by light microscopy, and a binding index (number of yeasts bound/50 macrophages/well or chamber) and a phagocytic index (number of yeasts phagocytosed/50 macrophages/well or chamber) was determined for each replicate in 96 wells or chamber slides (49). Each assay was repeated a minimum of three times. More than 30 different cell donors were used, and there was a remarkable consistency in the numbers of yeast cells phagocytosed per macrophage whether the cells were from male or female donors or from one week to the next with the same donor.

SAP and other additives.Human SAP or buffer-exchanged SAP (as described by Shao et al. [22]) in serum-free medium was added to wells to a final concentration of 30 µg/ml (the normal level in human serum). Other additives included CRP (Fitzgerald Industries, Acton, MA), D-mannose (Sigma-Aldrich), EDTA (Sigma-Aldrich), normal human male AB serum (Innovative Research), or bovine serum albumin (BSA) (Sigma-Aldrich) as described in the Results.

Flow cytometry.Fungi were cultured overnight (C. albicans) or for 48 h (S. cerevisiae) as described above, collected by centrifugation, and resuspended in TBS-C. Cells were incubated in undiluted normal human male AB serum or in TBS-C with 30 µg/ml SAP for 1 h at 4°C, washed three times by centrifugation, and resuspended in TBS-C. Cells were then incubated in TBS-C with 1:100 rabbit polyclonal anti-human SAP antibodies (PA5-24171; Invitrogen), or 1:100 anti-human CRP mouse monoclonal antibodies (MABF1070; clone 2A8.1, EMD-Millipore Corporation) for 30 min at 4°C. Yeasts were then washed three times and incubated with 1:200 fluorescein-labeled goat anti-rabbit or anti-mouse antibody (Invitrogen) for 30 min, washed three times, and resuspended in TBS. Fluorescence of cells was read on a BD LSR II flow cytometer (BD Biosciences, San Jose, CA) and then analyzed using Flowjo version 10.

Determination of macrophage antigen expression by immunocytochemistry.Macrophages were cultured in serum-free medium in 96-well plates as described above. After 5 days, the medium was replaced with the following: fresh serum-free medium for 30 or 60 min (control); serum-free medium plus C. albicans yeasts for 30 min; serum-free medium plus SAP (30 µg/ml) for 1 h; serum-free medium plus SAP (30 µg/ml) for 1 h followed by C. albicans yeasts for 30 min; or serum-free medium with C. albicans yeasts that had been soaked in normal human serum at 4°C for 30 min. For staining, macrophages were fixed with acetone for 15 min and air dried for 15 min, and nonspecific binding was blocked by incubation in PBS containing 4% BSA (PBS-BSA) for 60 min (22). Wells were then incubated with 5 µg/ml primary antibodies in PBS-BSA for 60 min as previously described (38). Isotype-matched irrelevant antibodies were used as controls. Antibodies from BioLegend (San Diego, CA) were anti-CD45, mouse IgG1, (catalog no. 304002), anti-CD54 (ICAM), mouse IgG1 (catalog no. 353102), and anti-CD206, mouse IgG1 (catalog no. 321102). Antibodies from Abcam (Cambridge, UK) were anti-CD209, mouse IgG1 (ab89186), anti-IRF5, rabbit monoclonal (EPR6094), and anti-IFR4, rabbit monoclonal (EP5699). Irrelevant mouse IgG1 (BioLegend) or rabbit polyclonal antibodies (R&D Systems, Minneapolis, MN) at 5 µg/ml in PBS-BSA were used as controls. Primary antibodies were detected with either biotinylated donkey F(ab′)2 anti-mouse IgG or biotinylated donkey F(ab′)2 anti-rabbit IgG (all cross-adsorbed against human Ig; Jackson ImmunoResearch, West Grove, PA). All secondary antibodies were used at 1 µg/ml in PBS-BSA for 30 min. Biotinylated antibodies were detected by a 1/500 dilution of ExtrAvidin alkaline phosphatase (Vector Laboratories, Burlingame, CA) in PBS-BSA. Staining was developed with the Vector Red Alkaline Phosphatase kit (Vector Laboratories) for 5 to 7 min and then counterstained with Gill’s hematoxylin number 3 (Sigma-Aldrich) following the manufacturer’s directions. Macrophages were identified as 15- to 40-µm-diameter cells with a large nucleus and pronounced cytoplasm. Cells were washed, dried, and later rehydrated with water, and cells were counted microscopically. There were six wells of macrophages for each antibody, and the experiment was repeated twice (100 macrophages were counted per well).

Polymorphonuclear leukocyte phagocytosis of yeasts.Blood samples from five healthy adult volunteers (three women and two men) were drawn into vacutainer tubes containing EDTA (Fisher Scientific, Waltham, MA) with written approval following a University of Arizona IRB-approved protocol. The blood was layered onto Polymorphoprep (Thermo Fisher) in a polypropylene tube and processed according to the manufacturer’s directions and centrifuged at 500 × g for 30 min. Isolated polymorphonuclear leukocytes (PMNs) were washed with TBS-C by centrifugation at 500 × g for 5 min twice and resuspended in TBS-C to 107cells/ml. Then 100 µl was placed in each well of 96-well plates, incubated with 30 µg/ml SAP or without SAP for 1 h at 37°C, and washed with buffer. Yeasts were added at a ratio of 100 to 10 yeasts/PMN or yeasts incubated in serum for 1 h were added to PMNs and incubated for 30 min at 37°C with shaking. The cells were washed three times with TBS-C and stained with Wright-Giemsa. The number of yeasts phagocytosed per 50 PMNs was determined by microscopy.

Cytokine measurements.Macrophages were cultured in serum-free medium as described above in 96-well plates. After 5 days, the medium was replaced with the following: fresh serum-free medium; serum-free medium plus 30 µg/ml SAP (1 h); serum-free medium with C. albicans yeasts; or serum free-medium plus SAP, followed by addition of yeasts for 30 min. Media were aspirated from the wells at various time points and stored at −20°C. Thawed media were assayed for human IL-6, TNF-α, IFN-γ, IL-10, and IL-17A homodimers using Ready-SET-Go ELISA kits following the manufacturer’s protocols (Invitrogen).

Statistics.All population totals were graphed using median values and 95% confidence intervals or means with standard errors. An unpaired, nonparametric Mann-Whitney test or a basic unpaired t test was used to determine statistical significance between different populations and treatment conditions. Medians are shown in some graphs to show the central tendency. Software used was Prism version 7 (Graphpad, San Diego, CA).

ACKNOWLEDGMENTS

We thank Robert Hershoff for his assistance with photomicroscopy and Jordan Bishop and Maria Love for their help with macrophage culture and experiments. We thank Nafees Ahmad for use of his laboratory for parts of this research. We also thank Joel Buxbaum and Marinos Kallikourdis for their expert work reviewing this work.

This work was supported by NIH grant HL118507.

FOOTNOTES

    • Received 25 January 2019
    • Accepted 30 January 2019
    • Published 12 March 2019
  • Copyright © 2019 Behrens et al.

This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

REFERENCES

  1. 1.↵
    1. Nelson SR,
    2. Tennent GA,
    3. Sethi D,
    4. Gower PE,
    5. Ballardie FW,
    6. Amatayakul-Chantler S,
    7. Pepys MB
    . 1991. Serum amyloid P component in chronic renal failure and dialysis. Clin Chim Acta 200:191–199. doi:10.1016/0009-8981(91)90090-Y.
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    1. Dillingh MR,
    2. van den Blink B,
    3. Moerland M,
    4. van Dongen MG,
    5. Levi M,
    6. Kleinjan A,
    7. Wijsenbeek MS,
    8. Lupher ML,
    9. Harper DM,
    10. Getsy JA,
    11. Hoogsteden HC,
    12. Burggraaf J
    . 2013. Recombinant human serum amyloid P in healthy volunteers and patients with pulmonary fibrosis. Pulm Pharmacol Ther 26:672–676. doi:10.1016/j.pupt.2013.01.008.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Herbert J,
    2. Hutchinson WL,
    3. Carr J,
    4. Ives J,
    5. Jakob-Roetne R,
    6. Yamamura K,
    7. Suzuki M,
    8. Pepys MB
    . 2002. Influenza virus infection is not affected by serum amyloid P component. Mol Med 8:9–15. doi:10.1007/BF03401998.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. de Haas CJ,
    2. van Leeuwen EM,
    3. van Bommel T,
    4. Verhoef J,
    5. van Kessel KP,
    6. van Strijp JA
    . 2000. Serum amyloid P component bound to Gram-negative bacteria prevents lipopolysaccharide-mediated classical pathway complement activation. Infect Immun 68:1753–1759. doi:10.1128/IAI.68.4.1753-1759.2000.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Noursadeghi M,
    2. Bickerstaff MC,
    3. Gallimore JR,
    4. Herbert J,
    5. Cohen J,
    6. Pepys MB
    . 2000. Role of serum amyloid P component in bacterial infection: protection of the host or protection of the pathogen. Proc Natl Acad Sci U S A 97:14584–14589. doi:10.1073/pnas.97.26.14584.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Pepys MB
    . 1994. Amyloidosis, p 637–655. In Frank MM, Austen KF, Claman HN, Unanue ER (ed), Samter's immunological diseases, ed 5. Little, Brown, Boston, MA.
  7. 7.↵
    1. Garcia M,
    2. Lipke P,
    3. Klotz S
    . 2013. Pathogenic microbial amyloids: their function and the host response. OA Microbiol 1:2.
    OpenUrlCrossRef
  8. 8.↵
    1. Lipke PN,
    2. Garcia MC,
    3. Alsteens D,
    4. Ramsook CB,
    5. Klotz SA,
    6. Dufrêne YF
    . 2012. Strengthening relationships: amyloids create adhesion nanodomains in yeasts. Trends Microbiol 20:59–65. doi:10.1016/j.tim.2011.10.002.
    OpenUrlCrossRefPubMedWeb of Science
  9. 9.↵
    1. Lipke PN,
    2. Klotz SA,
    3. Dufrene YF,
    4. Jackson DN,
    5. Garcia-Sherman MC
    . 2018. Amyloid-like β-aggregates as force-sensitive switches in fungal biofilms and infections. Microbiol Mol Biol Rev 82:e00035-17. doi:10.1128/MMBR.00035-17.
    OpenUrlCrossRef
  10. 10.↵
    1. Ramsook CB,
    2. Tan C,
    3. Garcia MC,
    4. Fung R,
    5. Soybelman G,
    6. Henry R,
    7. Litewka A,
    8. O'Meally S,
    9. Otoo HN,
    10. Khalaf RA,
    11. Dranginis AM,
    12. Gaur NK,
    13. Klotz SA,
    14. Rauceo JM,
    15. Jue CK,
    16. Lipke PN
    . 2010. Yeast cell adhesion molecules have functional amyloid-forming sequences. Eukaryot Cell 9:393–404. doi:10.1128/EC.00068-09.
    OpenUrlAbstract/FREE Full Text
  11. 11.↵
    1. Gilchrist KB,
    2. Garcia MC,
    3. Sobonya R,
    4. Lipke PN,
    5. Klotz SA
    . 2012. New features of invasive candidiasis in humans: amyloid formation by fungi and deposition of serum amyloid P component by the host. J Infect Dis 206:1473–1478. doi:10.1093/infdis/jis464.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Garcia-Sherman MC,
    2. Lundberg T,
    3. Sobonya RE,
    4. Lipke PN,
    5. Klotz SA
    . 2015. A unique biofilm in human deep mycoses: fungal amyloid is bound by host serum amyloid P component. NPJ Biofilms Microbiomes 1:15009. doi:10.1038/npjbiofilms.2015.9.
    OpenUrlCrossRef
  13. 13.↵
    1. Golconda U,
    2. Sobonya RE,
    3. Klotz SA
    . 2018. Do pentraxins bind to fungi in invasive human gastrointestinal candidiasis? J Fungi (Basel) 4:E111. doi:10.3390/jof4030111.
    OpenUrlCrossRef
  14. 14.↵
    1. Ben-Ami R,
    2. Luna M,
    3. Lewis RE,
    4. Walsh TJ,
    5. Kontoyiannis DP
    . 2009. A clinicopathological study of pulmonary mucormycosis in cancer patients: extensive angioinvasion but limited inflammatory response. J Infect 59:134–138. doi:10.1016/j.jinf.2009.06.002.
    OpenUrlCrossRefPubMedWeb of Science
  15. 15.↵
    1. Shaukat A,
    2. Bakri F,
    3. Young P,
    4. Hahn T,
    5. Ball D,
    6. Baer MR,
    7. Wetzler M,
    8. Slack JL,
    9. Loud P,
    10. Czuczman M,
    11. McCarthy PL,
    12. Walsh TJ,
    13. Segal BH
    . 2005. Invasive filamentous fungal infections in allogeneic hematopoietic stem cell transplant recipients after recovery from neutropenia: clinical, radiologic, and pathologic characteristics. Mycopathologia 159:181–188. doi:10.1007/s11046-004-5495-0.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    1. Stergiopoulou T,
    2. Meletiadis J,
    3. Roilides E,
    4. Kleiner DE,
    5. Schaufele R,
    6. Roden M,
    7. Harrington S,
    8. Dad L,
    9. Segal B,
    10. Walsh TJ
    . 2007. Host-dependent patterns of tissue injury in invasive pulmonary aspergillosis. Am J Clin Pathol 127:349–355. doi:10.1309/UJRV9DLC11RM3G8R.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Pepys MB
    . 2006. Amyloidosis. Annu Rev Med 57:223–241. doi:10.1146/annurev.med.57.121304.131243.
    OpenUrlCrossRefPubMedWeb of Science
  18. 18.↵
    1. Pepys MB,
    2. Dyck RF,
    3. de Beer FC,
    4. Skinner M,
    5. Cohen AS
    . 1979. Binding of serum amyloid P-component (SAP) by amyloid fibrils. Clin Exp Immunol 38:284–293.
    OpenUrlPubMedWeb of Science
  19. 19.↵
    1. Garcia MC,
    2. Lee JT,
    3. Ramsook CB,
    4. Alsteens D,
    5. Dufrêne YF,
    6. Lipke PN
    . 2011. A role for amyloid in cell aggregation and biofilm formation. PLoS One 6:e17632. doi:10.1371/journal.pone.0017632.
    OpenUrlCrossRef
  20. 20.↵
    1. Gaur NK,
    2. Klotz SA
    . 1997. Expression, cloning, and characterization of a Candida albicans gene, ALA1, that confers adherence properties upon Saccharomyces cerevisiae for extracellular matrix proteins. Infect Immun 65:5289–5294.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    1. Gaur NK,
    2. Klotz SA,
    3. Henderson RL
    . 1999. Overexpression of the Candida albicans ALA1 gene in Saccharomyces cerevisiae results in aggregation following attachment of yeast cells to extracellular matrix proteins, adherence properties similar to those of Candida albicans. Infect Immun 67:6040–6047.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    1. Shao DD,
    2. Suresh R,
    3. Vakil V,
    4. Gomer RH,
    5. Pilling D
    . 2008. Pivotal advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation. J Leukoc Biol 83:1323–1333. doi:10.1189/jlb.1107782.
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.↵
    1. Cox N,
    2. Pilling D,
    3. Gomer RH
    . 2014. Serum amyloid P: a systemic regulator of the innate immune response. J Leukoc Biol 96:739–743. doi:10.1189/jlb.1MR0114-068R.
    OpenUrlCrossRefPubMedWeb of Science
  24. 24.↵
    1. Siripont J,
    2. Tebo JM,
    3. Mortensen RF
    . 1988. Receptor-mediated binding of the acute-phase reactant mouse serum amyloid P-component (SAP) to macrophages. Cell Immunol 117:239–252. doi:10.1016/0008-8749(88)90115-3.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Murray PJ,
    2. Allen JE,
    3. Biswas SK,
    4. Fisher EA,
    5. Gilroy DW,
    6. Goerdt S,
    7. Gordon S,
    8. Hamilton JA,
    9. Ivashkiv LB,
    10. Lawrence T,
    11. Locati M,
    12. Mantovani A,
    13. Martinez FO,
    14. Mege JL,
    15. Mosser DM,
    16. Natoli G,
    17. Saeij JP,
    18. Schultze JL,
    19. Shirey KA,
    20. Sica A,
    21. Suttles J,
    22. Udalova I,
    23. van Ginderachter JA,
    24. Vogel SN,
    25. Wynn TA
    . 2014. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41:14–20. doi:10.1016/j.immuni.2014.06.008.
    OpenUrlCrossRefPubMedWeb of Science
  26. 26.↵
    1. Stahl PD,
    2. Ezekowitz RA
    . 1998. The mannose receptor is a pattern recognition receptor involved in host defense. Curr Opin Immunol 10:50–55. doi:10.1016/S0952-7915(98)80031-9.
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.↵
    1. Maródi L,
    2. Korchak HM,
    3. Johnston RB
    . 1991. Mechanisms of host defense against Candida species. I. Phagocytosis by monocytes and monocyte-derived macrophages. J Immunol 146:2783–2789.
    OpenUrlAbstract
  28. 28.↵
    1. Cambi A,
    2. Gijzen K,
    3. de Vries I,
    4. Torensma R,
    5. Joosten B,
    6. Adema GJ,
    7. Netea MG,
    8. Kullberg BJ,
    9. Romani L,
    10. Figdor CG
    . 2003. The C-type lectin DC-SIGN (CD209) is an antigen-uptake receptor for Candida albicans on dendritic cells. Eur J Immunol 33:532–538. doi:10.1002/immu.200310029.
    OpenUrlCrossRefPubMedWeb of Science
  29. 29.↵
    1. Chan CX,
    2. El-Kirat-Chatel S,
    3. Joseph IG,
    4. Jackson DN,
    5. Ramsook CB,
    6. Dufrêne YF,
    7. Lipke PN
    . 2016. Force sensitivity in Saccharomyces cerevisiae flocculins. mSphere 1:e00128-16. doi:10.1128/mSphere.00128-16.
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    1. Lipke PN,
    2. Ramsook C,
    3. Garcia-Sherman MC,
    4. Jackson DN,
    5. Chan CX,
    6. Bois M,
    7. Klotz SA
    . 2014. Between amyloids and aggregation lies a connection with strength and adhesion. New J Sci 2014:815102. doi:10.1155/2014/815102.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Van Gerven N,
    2. Van der Verren SE,
    3. Reiter DM,
    4. Remaut H
    . 2018. The role of functional amyloids in bacterial virulence. J Mol Biol 430:3657–3684. doi:10.1016/j.jmb.2018.07.010.
    OpenUrlCrossRef
  32. 32.↵
    1. Ramsugit S,
    2. Pillay M
    . 2014. Mycobacterium tuberculosis pili promote adhesion to and invasion of THP-1 macrophages. Jpn J Infect Dis 67:476–478. doi:10.7883/yoken.67.476.
    OpenUrlCrossRef
  33. 33.↵
    1. Kaur S,
    2. Singh PP
    . 2004. Serum amyloid P-component-mediated inhibition of the uptake of Mycobacterium tuberculosis by macrophages, in vitro. Scand J Immunol 59:425–431. doi:10.1111/j.0300-9475.2004.01412.x.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Sasada M,
    2. Johnston RB
    . 1980. Macrophage microbicidal activity. Correlation between phagocytosis-associated oxidative metabolism and the killing of Candida by macrophages. J Exp Med 152:85–98. doi:10.1084/jem.152.1.85.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    1. Krysan DJ,
    2. Sutterwala FS,
    3. Wellington M
    . 2014. Catching fire: Candida albicans, macrophages, and pyroptosis. PLoS Pathog 10:e1004139. doi:10.1371/journal.ppat.1004139.
    OpenUrlCrossRef
  36. 36.↵
    1. Xiang W,
    2. Cox N,
    3. Gomer RH
    . 2017. Identification of compounds that decrease numbers of Mycobacteria in human macrophages in the presence of serum amyloid P. J Leukoc Biol 102:857–869. doi:10.1189/jlb.1A0317-118RR.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Bharadwaj D,
    2. Mold C,
    3. Markham E,
    4. Du Clos TW
    . 2001. Serum amyloid P component binds to Fc gamma receptors and opsonizes particles for phagocytosis. J Immunol 166:6735–6741. doi:10.4049/jimmunol.166.11.6735.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Pilling D,
    2. Galvis-Carvajal E,
    3. Karhadkar TR,
    4. Cox N,
    5. Gomer RH
    . 2017. Monocyte differentiation and macrophage priming are regulated differentially by pentraxins and their ligands. BMC Immunol 18:30. doi:10.1186/s12865-017-0214-z.
    OpenUrlCrossRef
  39. 39.↵
    1. Alsteens D,
    2. Garcia MC,
    3. Lipke PN,
    4. Dufrêne YF
    . 2010. Force-induced formation and propagation of adhesion nanodomains in living fungal cells. Proc Natl Acad Sci U S A 107:20744–20749. doi:10.1073/pnas.1013893107.
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    1. Ballmann GE,
    2. Caffin WL
    . 1979. Lipid synthesis during reinitiation of growth from stationary phase cultures of Candida albicans. Mycopathologia 67:39–43. doi:10.1007/BF00436239.
    OpenUrlCrossRefPubMedWeb of Science
  41. 41.↵
    1. Orlean P
    . 2012. Architecture and biosynthesis of the Saccharomyces cerevisiae cell wall. Genetics 192:775–818. doi:10.1534/genetics.112.144485.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Christner RB,
    2. Mortensen RF
    . 1994. Binding of human serum amyloid P-component to phosphocholine. Arch Biochem Biophys 314:337–343. doi:10.1006/abbi.1994.1451.
    OpenUrlCrossRefPubMedWeb of Science
  43. 43.↵
    1. Bottazzi B,
    2. Santini L,
    3. Savino S,
    4. Giuliani MM,
    5. Dueñas Díez AI,
    6. Mancuso G,
    7. Beninati C,
    8. Sironi M,
    9. Valentino S,
    10. Deban L,
    11. Garlanda C,
    12. Teti G,
    13. Pizza M,
    14. Rappuoli R,
    15. Mantovani A
    . 2015. Recognition of Neisseria meningitidis by the long pentraxin PTX3 and its role as an endogenous adjuvant. PLoS One 10:e0120807. doi:10.1371/journal.pone.0120807.
    OpenUrlCrossRef
  44. 44.↵
    1. Cox N,
    2. Pilling D,
    3. Gomer RH
    . 2014. Distinct Fcγ receptors mediate the effect of serum amyloid P on neutrophil adhesion and fibrocyte differentiation. J Immunol 193:1701–1708. doi:10.4049/jimmunol.1400281.
    OpenUrlAbstract/FREE Full Text
  45. 45.↵
    1. Maharjan AS,
    2. Roife D,
    3. Brazill D,
    4. Gomer RH
    . 2013. Serum amyloid P inhibits granulocyte adhesion. Fibrogenesis Tissue Repair 6:2. doi:10.1186/1755-1536-6-2.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Cox N,
    2. Pilling D,
    3. Gomer RH
    . 2015. DC-SIGN activation mediates the differential effects of SAP and CRP on the innate immune system and inhibits fibrosis in mice. Proc Natl Acad Sci U S A 112:8385–8390. doi:10.1073/pnas.1500956112.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. van de Veerdonk FL,
    2. Marijnissen RJ,
    3. Kullberg BJ,
    4. Koenen HJ,
    5. Cheng SC,
    6. Joosten I,
    7. van den Berg WB,
    8. Williams DL,
    9. van der Meer JW,
    10. Joosten LA,
    11. Netea MG
    . 2009. The macrophage mannose receptor induces IL-17 in response to Candida albicans. Cell Host Microbe 5:329–340. doi:10.1016/j.chom.2009.02.006.
    OpenUrlCrossRefPubMedWeb of Science
  48. 48.↵
    1. Klotz SA,
    2. Sobonya RE,
    3. Lipke PN,
    4. Garcia-Sherman MC
    . 2016. Serum amyloid P component and systemic fungal infection: does it protect the host or is it a Trojan horse? Open Forum Infect Dis 3:ofw166. doi:10.1093/ofid/ofw166.
    OpenUrlCrossRef
  49. 49.↵
    1. Du C,
    2. Calderone RA
    . 2009. Phagocytosis and killing assays for Candida species. Methods Mol Biol 499:17–26. doi:10.1007/978-1-60327-151-6_3.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Download PDF
Citation Tools
Serum Amyloid P Component Binds Fungal Surface Amyloid and Decreases Human Macrophage Phagocytosis and Secretion of Inflammatory Cytokines
Nicole E. Behrens, Peter N. Lipke, Darrell Pilling, Richard H. Gomer, Stephen A. Klotz
mBio Mar 2019, 10 (2) e00218-19; DOI: 10.1128/mBio.00218-19

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this mBio article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Serum Amyloid P Component Binds Fungal Surface Amyloid and Decreases Human Macrophage Phagocytosis and Secretion of Inflammatory Cytokines
(Your Name) has forwarded a page to you from mBio
(Your Name) thought you would be interested in this article in mBio.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Serum Amyloid P Component Binds Fungal Surface Amyloid and Decreases Human Macrophage Phagocytosis and Secretion of Inflammatory Cytokines
Nicole E. Behrens, Peter N. Lipke, Darrell Pilling, Richard H. Gomer, Stephen A. Klotz
mBio Mar 2019, 10 (2) e00218-19; DOI: 10.1128/mBio.00218-19
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • INTRODUCTION
    • RESULTS
    • DISCUSSION
    • MATERIALS AND METHODS
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

SAP
cytokines
functional amyloid
innate immunity
macrophage polarization

Related Articles

Cited By...

About

  • About mBio
  • Editor in Chief
  • Board of Editors
  • AAM Fellows
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Author Warranty
  • Article Types
  • Ethics
  • Contact Us

Follow #mBio

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Online ISSN: 2150-7511