Genetic control of susceptibility to infection with Candida albicans in mice

Irena Radovanovic, Alaka Mullick, Philippe Gros, Irena Radovanovic, Alaka Mullick, Philippe Gros

Abstract

Candida albicans is an opportunistic pathogen that causes acute disseminated infections in immunocompromised hosts, representing an important cause of morbidity and mortality in these patients. To study the genetic control of susceptibility to disseminated C. albicans in mice, we phenotyped a group of 23 phylogenetically distant inbred strains for susceptibility to infection as measured by extent of fungal replication in the kidney 48 hours following infection. Susceptibility was strongly associated with the loss-of-function mutant complement component 5 (C5/Hc) allele, which is known to be inherited by approximately 40% of inbred strains. Our survey identified 2 discordant strains, AKR/J (C5-deficient, resistant) and SM/J (C5-sufficient, susceptible), suggesting that additional genetic effects may control response to systemic candidiasis in these strains. Haplotype association mapping in the 23 strains using high density SNP maps revealed several putative loci regulating the extent of C. albicans replication, amongst which the most significant were C5 (P value = 2.43×10(-11)) and a novel effect on distal chromosome 11 (P value = 7.63×10(-9)). Compared to other C5-deficient strains, infected AKR/J strain displays a reduced fungal burden in the brain, heart and kidney, and increased survival, concomitant with uniquely high levels of serum IFNγ. C5-independent genetic effects were further investigated by linkage analysis in an [A/JxAKR/J]F2 cross (n = 158) where the mutant Hc allele is fixed. These studies identified a chromosome 11 locus (Carg4, Candida albicans resistance gene 4; LOD = 4.59), and a chromosome 8 locus (Carg3; LOD = 3.95), both initially detected by haplotype association mapping. Alleles at both loci were inherited in a co-dominant manner. Our results verify the important effect of C5-deficiency in inbred mouse strains, and further identify two novel loci, Carg3 and Carg4, which regulate resistance to C. albicans infection in a C5-independent manner.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Kidney fungal burden, a measure…
Figure 1. Kidney fungal burden, a measure of susceptibility to C. albicans infection, in 23 inbred mouse strains.
23 out of 36 commonly used inbred strains from the Jackson Mouse Phenome Database were phenotyped for susceptibility to C. albicans. 7–10 female mice per strain were infected intravenously with 5×104C. albicans blastospores and the kidney fungal load was measured 48 h post-infection. Bars represent strain mean±SD. Horizontal lines represent mean (solid line) across all 23 strains±SD (dashed lines).
Figure 2. Genome-wide association mapping using EMMA.
Figure 2. Genome-wide association mapping using EMMA.
Using the R package implementation of EMMA, genome-wide association mapping was conducted across 23 inbred mouse strains. We obtained genotype data consisting of 132 000 SNPs from the Mouse HapMap project and rejected SNPs with MAF 10 transformed P values indicating association significance for each SNP genome-wide (A). Standardized threshold was set at P value 1.0×10−5 (solid line) and the Bonferroni multiple testing correction threshold was calculated to be 7.99×10−8 (dashed line). The most significant hits on chromosomes 2 and 11 are depicted (B and C) along with respective haplotype maps of susceptible (S) and resistant (R) strains at those locations. The conserved haplotype blocks are indicated with brackets, along with the Hc position (B) and the arrow showing the most conserved area (C). Asterisk denotes discordant strains with respect to the C5 genotype. Haplotype map colour coding: light gray (NA), gray (A/J like), and white (B6 like).
Figure 3. Phenotypic responses of discordant AKR/J…
Figure 3. Phenotypic responses of discordant AKR/J mouse strain upon C. albicans infection.
In 6 mice per strain, the heart (A), brain (B), and kidney (C) fungal load was measured 48 h after a low dose (5×104, i.v.) infection with C. albicans. In all organs, fungal load was significantly lower in AKR/J mice compared to A/J (**P<0.01, ***P<0.0001). Periodic acid-Schiff staining was performed on kidney sections (D) showing the extent of fungal burden and granulocyte infiltration. Magnification 100X and 400X (insert).
Figure 4. Differential susceptibility of A/J and…
Figure 4. Differential susceptibility of A/J and AKR/J mice to C. albicans infection.
6 mice per strain were challenged with the indicated dose of C. albicans and sacrificed at either 24 h or 48 h time point to determine the kidney fungal burden (A) and serum BUN levels (B). The fungal load was significantly lower in AKR/J mice compared to A/J, and was associated with lower BUN levels (*P<0.05, ***P≤0.0001, n.d. not done due to premature death of A/J mice). Bars represent group mean ± SD. For the survival study, 8 mice per strain were infected intravenously with a high dose (3×105) of C. albicans and monitored daily for clinical signs of morbidity (C). Survival curves were compared by a Log-rank test and the median survival of AKR/J (3 d) was found to be significantly different from A/J (1 d) (P<0.0001).
Figure 5. Inflammatory cytokine levels in the…
Figure 5. Inflammatory cytokine levels in the AKR/J mouse strain.
Serum cytokine levels in 3–6 mice of each strain were measured prior to infection and at the 48 h time point, as described in Materials and methods. Cytokine response of AKR/J mice resembled closely that of B6 strain and was significantly different from A/J mice. The standard deviation is indicated for each group. For all cytokines, infected AKR/J shows a statistically significant difference from infected A/J mice.
Figure 6. Linkage analysis in the informative…
Figure 6. Linkage analysis in the informative [A/JxAKR/J]F2 population.
[A/JxAKR/J]F2 mice (n = 158) were infected with C. albicans (5×104, i.v.) and kidneys were harvested 48 h post infection. CFU were determined as previously mentioned and results from four separate infections are plotted along with A/J, AKR/J, and [A/JxAKR/J]F1 controls (A). Each dot represents a single mouse. Distribution of kidney fungal load is shown in the entire [A/JxAKR/J]F2 population (B), after regression of log10CFU to an experiment and gender-specific mean (set at 0). Mice were genotyped at 257 SNPs and markers across the entire genome and interval mapping was carried out using the R/qtl software package. Whole genome LOD score traces are shown for genetic effects controlling kidney fungal burden in [A/JxAKR/J]F2 mice (C), identifying linkage to chromosome 8 (LOD = 3.95) and chromosome 11(LOD = 4.59), designated Carg3 and Carg4, respectively. Results for males and females are shown separately (dashed and dotted lines) and together (solid line), with marker positions on the x-axis. Permutation testing (1000 tests) identified genome-wide thresholds at P = 0.01 and 0.05.
Figure 7. Additive effect of the Carg3…
Figure 7. Additive effect of the Carg3 and Carg4 loci on kidney fungal burden in [A/JxAKR/J]F2 mice.
Detailed LOD score traces are shown for chromosome 11 (Carg4) (A) and chromosome 8 (Carg4) (B) loci for males, females, and both sexes combined. The shaded area designates the Bayesian 95% confidence interval for each locus and the genome-wide thresholds are indicated at P = 0.01 and P = 0.05. The additive effect of Carg3 and Carg4 loci is demonstrated by segregating the alleles of susceptible and resistant mice found at extremities (±1 standard deviation from the mean) of the distribution (C). Haplotype map colour coding: light gray (AKR/J), gray (A/J), and white (heterozygous). Each line represents a single mouse. Genome-wide association mapping results are depicted along with LOD score traces for chromosomes 11 (D) and 8 (E). The 10 Mb blocks examined for candidate genes are indicated for both loci.

References

    1. Mathews HLW-JL. CR, ed. Candida and candidiasis. Washington, DC.: ASM Press; 2002. Host defense against oral, esophageal, and gastrointestinal candidiasis. pp. 179–192.
    1. Spellberg BJ, Filler SG, Edwards JE., Jr Current treatment strategies for disseminated candidiasis. Clin Infect Dis. 2006;42:244–251.
    1. Pappas PG, Rex JH, Lee J, Hamill RJ, Larsen RA, et al. A prospective observational study of candidemia: epidemiology, therapy, and influences on mortality in hospitalized adult and pediatric patients. Clin Infect Dis. 2003;37:634–643.
    1. Morgan J. Global trends in candidemia: review of reports from 1995-2005. Curr Infect Dis Rep. 2005;7:429–439.
    1. Gudlaugsson O, Gillespie S, Lee K, Vande Berg J, Hu J, et al. Attributable mortality of nosocomial candidemia, revisited. Clin Infect Dis. 2003;37:1172–1177.
    1. Pfaller MA, Diekema DJ. Epidemiology of invasive candidiasis: a persistent public health problem. Clin Microbiol Rev. 2007;20:133–163.
    1. Shepherd MG, Poulter RT, Sullivan PA. Candida albicans: biology, genetics, and pathogenicity. Annu Rev Microbiol. 1985;39:579–614.
    1. Hector RF, Domer JE, Carrow EW. Immune responses to Candida albicans in genetically distinct mice. Infect Immun. 1982;38:1020–1028.
    1. Ashman RB, Bolitho EM, Papadimitriou JM. Patterns of resistance to Candida albicans in inbred mouse strains. Immunol Cell Biol. 1993;71(Pt 3):221–225.
    1. Lanza F. Clinical manifestation of myeloperoxidase deficiency. J Mol Med. 1998;76:676–681.
    1. Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB, et al. Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med. 2009;361:1760–1767.
    1. Puel A, Doffinger R, Natividad A, Chrabieh M, Barcenas-Morales G, et al. Autoantibodies against IL-17A, IL-17F, and IL-22 in patients with chronic mucocutaneous candidiasis and autoimmune polyendocrine syndrome type I. The Journal of experimental medicine. 2010;207:291–297.
    1. Glocker EO, Hennigs A, Nabavi M, Schaffer AA, Woellner C, et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med. 2009;361:1727–1735.
    1. Ashman RB. Protective and pathologic immune responses against Candida albicans infection. Front Biosci. 2008;13:3334–3351.
    1. Ashman RB, Fulurija A, Papadimitriou JM. Strain-dependent differences in host response to Candida albicans infection in mice are related to organ susceptibility and infectious load. Infect Immun. 1996;64:1866–1869.
    1. Ashman RB, Papadimitriou JM, Fulurija A, Drysdale KE, Farah CS, et al. Role of complement C5 and T lymphocytes in pathogenesis of disseminated and mucosal candidiasis in susceptible DBA/2 mice. Microb Pathog. 2003;34:103–113.
    1. Tuite A, Elias M, Picard S, Mullick A, Gros P. Genetic control of susceptibility to Candida albicans in susceptible A/J and resistant C57BL/6J mice. Genes Immun. 2005;6:672–682.
    1. Rhodes JC, Wicker LS, Urba WJ. Genetic control of susceptibility to Cryptococcus neoformans in mice. Infect Immun. 1980;29:494–499.
    1. Brummer E, Stevens DA. Anticryptococcal activity of macrophages: role of mouse strain, C5, contact, phagocytosis, and L-arginine. Cell Immunol. 1994;157:1–10.
    1. Mullick A, Elias M, Picard S, Bourget L, Jovcevski O, et al. Dysregulated inflammatory response to Candida albicans in a C5-deficient mouse strain. Infect Immun. 2004;72:5868–5876.
    1. Mullick A, Leon Z, Min-Oo G, Berghout J, Lo R, et al. Cardiac failure in C5-deficient A/J mice after Candida albicans infection. Infect Immun. 2006;74:4439–4451.
    1. Tuite A, Mullick A, Gros P. Genetic analysis of innate immunity in resistance to Candida albicans. Genes Immun. 2004;5:576–587.
    1. Lambris JD, Ricklin D, Geisbrecht BV. Complement evasion by human pathogens. Nat Rev Microbiol. 2008;6:132–142.
    1. Zipfel PF. Complement and immune defense: from innate immunity to human diseases. Immunol Lett. 2009;126:1–7.
    1. Tsoni SV, Kerrigan AM, Marakalala MJ, Srinivasan N, Duffield M, et al. Complement C3 plays an essential role in the control of opportunistic fungal infections. Infect Immun. 2009;77:3679–3685.
    1. Ashman RB. A gene (Carg1) that regulates tissue resistance to Candida albicans maps to chromosome 14 of the mouse. Microb Pathog. 1998;25:333–335.
    1. Ashman RB, Fulurija A, Papadimitriou JM. A second Candida albicans resistance gene (Carg2) regulates tissue damage, but not fungal clearance, in sub-lethal murine systemic infection. Microb Pathog. 1998;25:349–352.
    1. Wetsel RA, Fleischer DT, Haviland DL. Deficiency of the murine fifth complement component (C5). A 2-base pair gene deletion in a 5′-exon. J Biol Chem. 1990;265:2435–2440.
    1. Kang HM, Zaitlen NA, Wade CM, Kirby A, Heckerman D, et al. Efficient control of population structure in model organism association mapping. Genetics. 2008;178:1709–1723.
    1. Cheverud JM. A simple correction for multiple comparisons in interval mapping genome scans. Heredity. 2001;87:52–58.
    1. Frazer KA, Eskin E, Kang HM, Bogue MA, Hinds DA, et al. A sequence-based variation map of 8.27 million SNPs in inbred mouse strains. Nature. 2007;448:1050–1053.
    1. MacCallum DM, Castillo L, Brown AJ, Gow NA, Odds FC. Early-expressed chemokines predict kidney immunopathology in experimental disseminated Candida albicans infections. PLoS One. 2009;4:e6420.
    1. Romani L, Puccetti P, Mencacci A, Cenci E, Spaccapelo R, et al. Neutralization of IL-10 up-regulates nitric oxide production and protects susceptible mice from challenge with Candida albicans. J Immunol. 1994;152:3514–3521.
    1. Vazquez-Torres A, Jones-Carson J, Wagner RD, Warner T, Balish E. Early resistance of interleukin-10 knockout mice to acute systemic candidiasis. Infect Immun. 1999;67:670–674.
    1. Balish E, Wagner RD, Vazquez-Torres A, Pierson C, Warner T. Candidiasis in interferon-gamma knockout (IFN-gamma-/-) mice. J Infect Dis. 1998;178:478–487.
    1. Ashman RB, Fulurija A, Papadimitriou JM. Evidence that two independent host genes influence the severity of tissue damage and susceptibility to acute pyelonephritis in murine systemic candidiasis. Microb Pathog. 1997;22:187–192.
    1. Kitsios GD, Tangri N, Castaldi PJ, Ioannidis JP. Laboratory mouse models for the human genome-wide associations. PLoS One. 2010;5:e13782.
    1. Kirby A, Kang HM, Wade CM, Cotsapas C, Kostem E, et al. Fine mapping in 94 inbred mouse strains using a high-density haplotype resource. Genetics. 2010;185:1081–1095.
    1. Su WL, Sieberts SK, Kleinhanz RR, Lux K, Millstein J, et al. Assessing the prospects of genome-wide association studies performed in inbred mice. Mamm Genome. 2010;21:143–152.
    1. McClurg P, Pletcher MT, Wiltshire T, Su AI. Comparative analysis of haplotype association mapping algorithms. BMC Bioinformatics. 2006;7:61.
    1. Manenti G, Galvan A, Pettinicchio A, Trincucci G, Spada E, et al. Mouse genome-wide association mapping needs linkage analysis to avoid false-positive Loci. PLoS Genet. 2009;5:e1000331.
    1. Cenci E, Mencacci A, Del Sero G, d'Ostiani CF, Mosci P, et al. IFN-gamma is required for IL-12 responsiveness in mice with Candida albicans infection. J Immunol. 1998;161:3543–3550.
    1. Kullberg BJ, van 't Wout JW, Hoogstraten C, van Furth R. Recombinant interferon-gamma enhances resistance to acute disseminated Candida albicans infection in mice. J Infect Dis. 1993;168:436–443.
    1. Djeu JY, Blanchard DK, Halkias D, Friedman H. Growth inhibition of Candida albicans by human polymorphonuclear neutrophils: activation by interferon-gamma and tumor necrosis factor. J Immunol. 1986;137:2980–2984.
    1. Marodi L, Schreiber S, Anderson DC, MacDermott RP, Korchak HM, et al. Enhancement of macrophage candidacidal activity by interferon-gamma. Increased phagocytosis, killing, and calcium signal mediated by a decreased number of mannose receptors. J Clin Invest. 1993;91:2596–2601.
    1. Cohen MS, Isturiz RE, Malech HL, Root RK, Wilfert CM, et al. Fungal infection in chronic granulomatous disease. The importance of the phagocyte in defense against fungi. Am J Med. 1981;71:59–66.
    1. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75:163–189.
    1. Brierley MM, Fish EN. Stats: multifaceted regulators of transcription. J Interferon Cytokine Res. 2005;25:733–744.
    1. Robertson G, Hirst M, Bainbridge M, Bilenky M, Zhao Y, et al. Genome-wide profiles of STAT1 DNA association using chromatin immunoprecipitation and massively parallel sequencing. Nat Methods. 2007;4:651–657.
    1. Seifert U, Bialy LP, Ebstein F, Bech-Otschir D, Voigt A, et al. Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell. 2010;142:613–624.
    1. Duan B, Davis R, Sadat EL, Collins J, Sternweis PC, et al. Distinct roles of adenylyl cyclase VII in regulating the immune responses in mice. J Immunol. 2010;185:335–344.
    1. Watson PA, Krupinski J, Kempinski AM, Frankenfield CD. Molecular cloning and characterization of the type VII isoform of mammalian adenylyl cyclase expressed widely in mouse tissues and in S49 mouse lymphoma cells. J Biol Chem. 1994;269:28893–28898.
    1. Ernst PB, Garrison JC, Thompson LF. Much ado about adenosine: adenosine synthesis and function in regulatory T cell biology. J Immunol. 2010;185:1993–1998.
    1. Hertz AL, Bender AT, Smith KC, Gilchrist M, Amieux PS, et al. Elevated cyclic AMP and PDE4 inhibition induce chemokine expression in human monocyte-derived macrophages. Proc Natl Acad Sci U S A. 2009;106:21978–21983.
    1. Peters-Golden M. Putting on the brakes: cyclic AMP as a multipronged controller of macrophage function. Sci Signal. 2009;2:pe37.
    1. Buhl AM, Avdi N, Worthen GS, Johnson GL. Mapping of the C5a receptor signal transduction network in human neutrophils. Proc Natl Acad Sci U S A. 1994;91:9190–9194.
    1. Choubey D, Panchanathan R. Interferon-inducible Ifi200-family genes in systemic lupus erythematosus. Immunol Lett. 2008;119:32–41.
    1. Johnatty SE, Beesley J, Chen X, Macgregor S, Duffy DL, et al. Evaluation of candidate stromal epithelial cross-talk genes identifies association between risk of serous ovarian cancer and TERT, a cancer susceptibility "hot-spot". PLoS Genet. 2010;6:e1001016.
    1. Tan J, Qiao W, Wang J, Xu F, Li Y, et al. IFP35 is involved in the antiviral function of interferon by association with the viral tas transactivator of bovine foamy virus. J Virol. 2008;82:4275–4283.
    1. Bange FC, Vogel U, Flohr T, Kiekenbeck M, Denecke B, et al. IFP 35 is an interferon-induced leucine zipper protein that undergoes interferon-regulated cellular redistribution. J Biol Chem. 1994;269:1091–1098.
    1. Zhang L, Tang Y, Tie Y, Tian C, Wang J, et al. The PH domain containing protein CKIP-1 binds to IFP35 and Nmi and is involved in cytokine signaling. Cell Signal. 2007;19:932–944.
    1. Wang X, Johansen LM, Tae HJ, Taparowsky EJ. IFP 35 forms complexes with B-ATF, a member of the AP1 family of transcription factors. Biochem Biophys Res Commun. 1996;229:316–322.
    1. Schraml BU, Hildner K, Ise W, Lee WL, Smith WA, et al. The AP-1 transcription factor Batf controls T(H)17 differentiation. Nature. 2009;460:405–409.
    1. Saijo S, Ikeda S, Yamabe K, Kakuta S, Ishigame H, et al. Dectin-2 recognition of alpha-mannans and induction of Th17 cell differentiation is essential for host defense against Candida albicans. Immunity. 2010;32:681–691.
    1. Eyerich K, Foerster S, Rombold S, Seidl HP, Behrendt H, et al. Patients with chronic mucocutaneous candidiasis exhibit reduced production of Th17-associated cytokines IL-17 and IL-22. J Invest Dermatol. 2008;128:2640–2645.
    1. Kisand K, Boe Wolff AS, Podkrajsek KT, Tserel L, Link M, et al. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines. The Journal of experimental medicine. 2010;207:299–308.
    1. Zeller R. Fixation, embedding, and sectioning of tissues, embryos, and single cells. 2001;11 Current protocols in molecular biology/edited by Frederick M Ausubel, et al. Chapter 14: Unit 14.
    1. Fortin A, Diez E, Rochefort D, Laroche L, Malo D, et al. Recombinant congenic strains derived from A/J and C57BL/6J: a tool for genetic dissection of complex traits. Genomics. 2001;74:21–35.
    1. Haley CS, Knott SA. A simple regression method for mapping quantitative trait loci in line crosses using flanking markers. Heredity. 1992;69:315–324.
    1. Lander ES, Botstein D. Mapping mendelian factors underlying quantitative traits using RFLP linkage maps. Genetics. 1989;121:185–199.
    1. Broman KW, Wu H, Sen S, Churchill GA. R/qtl: QTL mapping in experimental crosses. Bioinformatics. 2003;19:889–890.

Source: PubMed

3
購読する