The altered landscape of the human skin microbiome in patients with primary immunodeficiencies

Julia Oh, Alexandra F Freeman, NISC Comparative Sequencing Program, Morgan Park, Robert Sokolic, Fabio Candotti, Steven M Holland, Julia A Segre, Heidi H Kong, Julia Oh, Alexandra F Freeman, NISC Comparative Sequencing Program, Morgan Park, Robert Sokolic, Fabio Candotti, Steven M Holland, Julia A Segre, Heidi H Kong

Abstract

While landmark studies have shown that microbiota activate and educate host immunity, how immune systems shape microbiomes and contribute to disease is incompletely characterized. Primary immunodeficiency (PID) patients suffer recurrent microbial infections, providing a unique opportunity to address this issue. To investigate the potential influence of host immunity on the skin microbiome, we examined skin microbiomes in patients with rare monogenic PIDs: hyper-IgE (STAT3-deficient), Wiskott-Aldrich, and dedicator of cytokinesis 8 syndromes. While specific immunologic defects differ, a shared hallmark is atopic dermatitis (AD)-like eczema. We compared bacterial and fungal skin microbiomes (41 PID, 13 AD, 49 healthy controls) at four clinically relevant sites representing the major skin microenvironments. PID skin displayed increased ecological permissiveness with altered population structures, decreased site specificity and temporal stability, and colonization with microbial species not observed in controls, including Clostridium species and Serratia marcescens. Elevated fungal diversity and increased representation of opportunistic fungi (Candida, Aspergillus) supported increased PID skin permissiveness, suggesting that skin may serve as a reservoir for the recurrent fungal infections observed in these patients. The overarching theme of increased ecological permissiveness in PID skin was counterbalanced by the maintenance of a phylum barrier in which colonization remained restricted to typical human-associated phyla. Clinical parameters, including markers of disease severity, were positively correlated with prevalence of Staphylococcus, Corynebacterium, and other less abundant taxa. This study examines differences in microbial colonization and community stability in PID skin and informs our understanding of host-microbiome interactions, suggesting a bidirectional dialogue between skin commensals and the host organism.

Figures

Figure 1.
Figure 1.
Representative clinical images of disease severity in the different patient groups. (A) Non-PID atopic dermatitis, (B) Hyper IgE syndrome, (C) Wiskott-Aldrich, and (D) DOCK8 deficiency.
Figure 2.
Figure 2.
Bacterial taxonomic classifications show colonization with unique taxa and altered representation of diverse taxa. Relative abundances of 14 major phyla-family taxonomies in the antecubital fossa (Af) are shown in A and retroauricular crease (Ra) in B. Shown are seven representative healthy controls and all Sanger-sequenced STAT3-HIES patients. Full versions of all skin and nares sites are in Supplemental Figure S4. (C) Inlaid pie charts compare the mean relative abundances of major skin genera across patient groups. Mean ± SEM for C are shown in Supplemental Figure S5 and Supplemental Tables S9 and S10. Patient identifiers are in Supplemental Table S13.
Figure 3.
Figure 3.
Taxonomies associated with variation within and between individuals. Principal coordinates analysis (PCoA) of the Yue-Clayton theta coefficient, which calculates the similarity between two samples based on (1) number of species in common between two samples, and (2) their relative abundances. Samples that have similar principal coordinates appear closer together, i.e., are more similar. Biplot lines indicate the most significant unique consensus taxonomies contributing to variation along axis 1; Spearman correlations (ρ) are with axis 1. Length of biplot lines reflects the contribution of that taxa to the top three axes. Associated P-values < 2.2 × 10−16. Percentage variation attributed to an axis is indicated. Antecubital fossa (Af) and retroauricular crease (Ra) are shown in A and B, respectively; nares and volar forearm are shown in Supplemental Figure S6.
Figure 4.
Figure 4.
Community-wide metrics suggest altered permissivity in niche specificity in colonization in PID individuals. Site codes: (Af) antecubital fossa; (N) nares; (Ra) retroauricular crease; (Vf) volar forearm. (A) Mean ± SEM for Shannon diversity is plotted for each patient group (colored as indicated) at all sites. (B) Site specificity is measured by the Yue-Clayton theta coefficient as in Figure 3, calculated between moist (antecubital fossa), dry (volar forearm), and sebaceous (retroauricular crease) skin sites and nares. (C) Longitudinal stability was assessed by calculating the theta coefficient between two and four timepoints for controls (light blue) and STAT3-HIES patients (red).
Figure 5.
Figure 5.
Taxonomic association with clinical features. (A) Pie charts comparing the mean relative representation of major staphylococcal species across patient groups at all sites. Site codes: (Af) antecubital fossa; (N) nares; (Ra) retroauricular crease; (Vf) volar forearm. (B) Correlation of taxonomy with clinical markers of disease severity for the antecubital fossa. Unsupervised hierarchical clustering of Spearman correlation coefficients for the 34 taxa whose mean abundance was >0.25% across STAT3-HIES patients. Correlations are calculated against blood levels of lactate dehydrogenase, lymphocytes, IgE, and eosinophils, and a score of skin disease severity (scoring atopic dermatitis [SCORAD]). Hemoglobin was a control. Reds indicate correlation; blues indicate anti-correlation.
Figure 6.
Figure 6.
Characterization of fungal skin communities suggest increased permissivity and elevated levels of opportunistic pathogenic fungi. (A) Boxplots showing fungal richness (species observed) of STAT3-HIES versus controls at all sites; black bars indicate median. (B) Select ITS1 fungal taxonomic classifications for the antecubital fossa, grouped by patient category. Full versions of all skin and nares sites are shown in Supplemental Figure S8. Patient identifiers are in Supplemental Table S13.

References

    1. Balter M 2012. Taking stock of the human microbiome and disease. Science 336: 1246–1247
    1. Benjamini Y, Hochberg Y 1995. Controlling the false discovery rate: A practical and powerful approach to multiple testing. J R Stat Soc Ser B Methodol 57: 289–300
    1. Berglund C, Söderquist B 2008. The origin of a methicillin-resistant Staphylococcus aureus isolate at a neonatal ward in Sweden: Possible horizontal transfer of a staphylococcal cassette chromosome mec between methicillin-resistant Staphylococcus haemolyticus and Staphylococcus aureus. Clin Microbiol Infect 14: 1048–1056
    1. Cho S-H, Strickland I, Boguniewicz M, Leung DYM 2001. Fibronectin and fibrinogen contribute to the enhanced binding of Staphylococcus aureus to atopic skin. J Allergy Clin Immunol 108: 269–274
    1. Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, Reading NC, Villablanca EJ, Wang S, Mora JR, et al. 2012. Gut immune maturation depends on colonization with a host-specific microbiota. Cell 149: 1578–1593
    1. Clayton MK, Yue JC 2005. A similarity measure based on species proportions. Commun Statist-Theor Method 34: 2123–2131
    1. Conlan S, Kong HH, Segre JA 2012. Species-level analysis of DNA sequence data from the NIH Human Microbiome Project. PLoS ONE 7: e47075.
    1. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R 2009. Bacterial community variation in human body habitats across space and time. Science 326: 1694–1697
    1. Denef VJ, Mueller RS, Banfield JF 2010. AMD biofilms: Using model communities to study microbial evolution and ecological complexity in nature. ISME J 4: 599–610
    1. DeSantis TZ, Hugenholtz P, Keller K, Brodie EL, Larsen N, Piceno YM, Phan R, Andersen GL 2006. NAST: A multiple sequence alignment server for comparative analysis of 16S rRNA genes. Nucleic Acids Res 34: W394–W399
    1. Dethlefsen L, McFall-Ngai M, Relman DA 2007. An ecological and evolutionary perspective on human–microbe mutualism and disease. Nature 449: 811–818
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R 2011. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27: 2194–2200
    1. Engelhardt KR, McGhee S, Winkler S, Sassi A, Woellner C, Lopez-Herrera G, Chen A, Kim HS, Lloret MG, Schulze I, et al. 2009. Large deletions and point mutations involving DOCK8 in the autosomal recessive form of the hyper-IgE syndrome. J Allergy Clin Immunol 124: 1289.
    1. Findley K, Oh J, Yang J, Conlan S, Deming C, Meyer JA, Schoenfeld D, Nomicos E, Park M, NIH Intramural Sequencing Center Comparative Sequencing Program, et al. 2013. Topographic diversity of fungal and bacterial communities in human skin. Nature 498: 367–370
    1. Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, NISC Comparative Sequencing Program, Bouffard GG, Blakesley RW, Murray PR, et al. 2009. Topographical and temporal diversity of the human skin microbiome. Science 324: 1190–1192
    1. Harris JK, Caporaso JG, Walker JJ, Spear JR, Gold NJ, Robertson CE, Hugenholtz P, Goodrich J, McDonald D, Knights D, et al. 2013. Phylogenetic stratigraphy in the Guerrero Negro hypersaline microbial mat. ISME J 7: 50–60
    1. Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, Brodsky N, Freeman AF, Demidowich A, Davis J, Turner ML, et al. 2007. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med 357: 1608–1619
    1. The Human Microbiome Project Consortium 2012. Structure, function and diversity of the healthy human microbiome. Nature 486: 207–214
    1. Jiang X, Clark RA, Liu L, Wagers AJ, Fuhlbrigge RC, Kupper TS 2012. Skin infection generates non-migratory memory CD8+ TRM cells providing global skin immunity. Nature 483: 227–231
    1. Knights D, Costello EK, Knight R 2011. Supervised classification of human microbiota. FEMS Microbiol Rev 35: 343–359
    1. Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, Nomicos E, Polley EC, Komarow HD, Murray PR, et al. 2012. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res 22: 850–859
    1. Kunz B, Oranje AP, Labrèze L, Stalder JF, Ring J, Taïeb A 1997. Clinical validation and guidelines for the SCORAD index: Consensus report of the European Task Force on Atopic Dermatitis. Dermatology (Basel) 195: 10–19
    1. Lai Y, Villaruz AE, Li M, Cha DJ, Sturdevant DE, Otto M 2007. The human anionic antimicrobial peptide dermcidin induces proteolytic defence mechanisms in staphylococci. Mol Microbiol 63: 497–506
    1. Lennon NJ, Lintner RE, Anderson S, Alvarez P, Barry A, Brockman W, Daza R, Erlich RL, Giannoukos G, Green L, et al. 2010. A scalable, fully automated process for construction of sequence-ready barcoded libraries for 454. Genome Biol 11: R15.
    1. Matsen F, Kodner R, Armbrust EV 2010. pplacer: Linear time maximum-likelihood and Bayesian phylogenetic placement of sequences onto a fixed reference tree. BMC Bioinformatics 11: 538.
    1. McCrea KW, Hartford O, Davis S, Eidhin DN, Lina G, Speziale P, Foster TJ, Höök M 2000. The serine-aspartate repeat (Sdr) protein family in Staphylococcus epidermidis. Microbiology 146: 1535–1546
    1. McGhee SA, Chatila TA 2010. DOCK8 immune deficiency as a model for primary cytoskeletal dysfunction. Dis Markers 29: 151–156
    1. Milner JD, Sandler NG, Douek DC 2010. Th17 cells, Job's syndrome and HIV: Opportunities for bacterial and fungal infections. Curr Opin HIV AIDS 5: 179–183
    1. Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, Kawamura N, Ariga T, Pasic S, Stojkovic O, et al. 2007. Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature 448: 1058–1062
    1. Naik S, Bouladoux N, Wilhelm C, Molloy MJ, Salcedo R, Kastenmuller W, Deming C, Quinones M, Koo L, Conlan S, et al. 2012. Compartmentalized control of skin immunity by resident commensals. Science 337: 1115–1119
    1. Nilsson M, Frykberg L, Flock J-I, Pei L, Lindberg M, Guss B 1998. A Fibrinogen-binding protein of Staphylococcus epidermidis. Infect Immun 66: 2666–2673
    1. Ochs HD, Thrasher AJ 2006. The Wiskott-Aldrich syndrome. J Allergy Clin Immunol 117: 725–738
    1. Oh J, Conlan S, Polley EC, Segre JA, Kong HH 2012. Shifts in human skin and nares microbiota of healthy children and adults. Genome Medicine 4: 77.
    1. Oranje AP, Glazenburg EJ, Wolkerstorfer A, de Waard-van der Spek FB 2007. Practical issues on interpretation of scoring atopic dermatitis: The SCORAD index, objective SCORAD and the three-item severity score. Br J Dermatol 157: 645–648
    1. Peschel A, Jack RW, Otto M, Collins LV, Staubitz P, Nicholson G, Kalbacher H, Nieuwenhuizen WF, Jung G, Tarkowski A, et al. 2001. Staphylococcus aureus resistance to human defensins and evasion of neutrophil killing via the novel virulence factor Mprf is based on modification of membrane lipids with l-lysine. J Exp Med 193: 1067–1076
    1. Peters BM, Noverr MC 2013. Candida albicans-Staphylococcus aureus polymicrobial peritonitis modulates host innate immunity. Infect Immun 81: 2178–2189
    1. Pruesse E, Quast C, Knittel K, Fuchs BM, Ludwig W, Peplies J, Glöckner FO 2007. SILVA: A comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res 35: 7188–7196
    1. Quince C, Lanzen A, Davenport R, Turnbaugh P 2011. Removing noise from pyrosequenced amplicons. BMC Bioinformatics 12: 38.
    1. Renner ED, Puck JM, Holland SM, Schmitt M, Weiss M, Frosch M, Bergmann M, Davis J, Belohradsky BH, Grimbacher B 2004. Autosomal recessive hyperimmunoglobulin E syndrome: a distinct disease entity. J Pediatr 144: 93–99
    1. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, Lesniewski RA, Oakley BB, Parks DH, Robinson CJ, et al. 2009. Introducing mothur: Open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol 75: 7537–7541
    1. Sieprawska-Lupa M, Mydel P, Krawczyk K, Wojcik K, Puklo M, Lupa B, Suder P, Silberring J, Reed M, Pohl J, et al. 2004. Degradation of human antimicrobial peptide LL-37 by Staphylococcus aureus-derived proteinases. Antimicrob Agents Chemother 48: 4673–4679
    1. Stone L, Roberts A 1990. The checkerboard score and species distributions. Oecologia 85: 74–79
    1. Upadhyay V, Poroyko V, Kim T, Devkota S, Fu S, Liu D, Tumanov AV, Koroleva EP, Deng L, Nagler C, et al. 2012. Lymphotoxin regulates commensal responses to enable diet-induced obesity. Nat Immunol 13: 947–953
    1. von Eiff C, Becker K, Machka K, Stammer H, Peters G 2001. Nasal carriage as a source of Staphylococcus aureus bacteremia. N Engl J Med 344: 11–16
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR 2007. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol 73: 5261–5267
    1. Wielders C, Vriens M, Brisse S, de Graaf-Miltenburg L, Troelstra A, Fleer A, Schmitz F, Verhoef J, Fluit A 2001. Evidence for in-vivo transfer of mecA DNA between strains of Staphylococcus aureus. Lancet 357: 1674–1675
    1. Williams HC, Burney PG, Pembroke AC, Hay RJ 1994. The U.K. Working Party's diagnostic criteria for atopic dermatitis. III. Independent hospital validation. Br J Dermatol 131: 406–416
    1. Williams HC, Burney PG, Pembroke AC, Hay RJ 1996. Validation of the U.K. diagnostic criteria for atopic dermatitis in a population setting. U.K. Diagnostic Criteria for Atopic Dermatitis Working Party. Br J Dermatol 135: 12–17
    1. Williams RJ, Henderson B, Sharp LJ, Nair SP 2002. Identification of a fibronectin-binding protein from Staphylococcus epidermidis. Infect Immun 70: 6805–6810
    1. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al. 2012. Human gut microbiome viewed across age and geography. Nature 486: 222–227
    1. Zhang Q, Davis JC, Lamborn IT, Freeman AF, Jing H, Favreau AJ, Matthews HF, Davis J, Turner ML, Uzel G, et al. 2009. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med 361: 2046–2055

Source: PubMed

3
Se inscrever