Journal List > Allergy Asthma Respir Dis > v.4(6) > 1059215

Kim, Suh, Lee, Kim, Cho, and Cho: Microbiome research in food allergy and atopic dermatitis

Abstract

Food allergy (FA) and atopic dermatitis (AD) are representative allergic diseases that begin early in life and result in considerable socioeconomic burden. While the pathophysiology and the optimal treatment modalities of these diseases are largely unknown, the role of microbes in health and disease are being highlighted. Recent advances in analyzing microbiome have enabled us to expand our research on impacts of the microbiome on the onset and course of FA and AD. Risk factors that are presumed to affect intestinal microbiome also modulate the onset of allergic diseases, which is more evident in AD than in FA. Considering animal studies, intestinal microbiota interacts with FA and the influence is bi-directional. The activation of regulatory T cell and the innate immune system is supposed to mediate the interaction. Regarding human studies, there exists the difference in the composition of microbiome between subjects with FA or AD and matched normal controls, which can further play as a predictive marker for later development of FA or AD. Probiotics are now investigated as a primary therapeutic agent or as an adjuvant tool for conventional therapies in preventing or modulating FA or AD. Currently, several reports on favorable outcomes become available, which should be replicated and backed up by large-scale studies with more detailed protocols.

Figures and Tables

Table 1

List of human microbiome studies on food allergy

aard-4-389-i001
Researcher Disease Subjects Design Sample Method Key findings Comments
Apostolou et al. (2001)32 Atopic dermatitis Adults 8 (sensitive) 9 (control) Case-control Feces FISH probing LGG-consumption resulted in Bifidobacteria (▲) in healthy but not in milk-sensitive subjects, as well as general (▲) in bacterial numbers. Profile, therapeutic effects
Thompson-Chagoyan et al. (2010)27 Cows milk allergy Infants 46 (allergic) 46 (control) Clinical trial Feces FISH probing Comparison of faecal samples from cows milk protein allergic infants (baseline/ 6 months) showed count and proportion of Lactobacilli (▲), counts and proportions of Enterobacteria (▼) and Bifidobacteria (▼). Profile longitudinal
Thompson-Chagoyan et al. (2011)73 Cows milk allergy Infants 46 (allergic) 46 (control) Case-control Feces FISH probing Milk-allergic infant faeces had Clostridium cocoides group (▲), Atopobium cluster (▲), and sum of proportions of the different bacterial groups (▲). Profile mechanisms
Ling et al. (2014)31 Food allergy Infants 17 (IgE-mediated) 17 (non-IgE-mediated) 45 (control) Case-control Feces 16S rRNA pyrosequencing Infants with IgE-mediated food allergy had Clotridium sensu stricto (▲), Anaerobacter (▲), and Bacteroides (▼), Clostridium XVIII (▼). Profile
Azad et al. (2015)29 Child cohort participants Infants 12 (sensitized) 154 (control) Case-control Feces 16S rRNA next generation sequencing Low gut microbiota richness and an elevated Enterobacteriaceae to Bacteroidanceae ratio in early infancy are linked with subsequent food sensitization. Profile longitudinal
Tang et al. (2015)33 Peanut allergy Children 31 (case) 31 (placebo) Observative N/A N/A Probiotics and peanut oral immunotherapy has good efficacy. Therapeutic effects
Chen et al. (2016)74 Food sensitization Children 23 (sensitized), 22 (control) Cohort Feces 16S rRNA pyrosequencing In sensitized groups, the number of Bacteroidetes (▼) and that of Firmicutes (▲). Profile
Hua et al. (2016)30 American gut project participants Adults 1,879 Case-control Feces 16S rRNA sequencing American adults with allergies have diversity (▼), Clostridiales (▼), and Bacteroidales (▲) in their gut microbiota. Profile
Berni Canani et al. (2016)28 Cows milk allergy Infants 19 (allergic) 20 (control) Clinical trial Feces 16S rRNA sequencing EHCF+LGG promote tolerance in milk allergic infants, in part, by influencing the strain-level bacterial community structure. Profile, therapeutic effects

FISH, fluorescence in situ hybridization; EHCF, extensively hydrolyzed casein formula; LGG, Lactobacillus GG; N/A, not applicable; ▲, increase; ▼, decrease.

Table 2

List of human microbiome studies on atopic dermatitis

aard-4-389-i002
Source Disease Subejcts Design Sample Method Key findings Comments
Dekio et al. (2007)49 AD Adults 13 (case) 10 (control) Case-control Skin swab Terminal restriction fragment length polymorphism (T-RFLP) analysis of 16S rRNA genes Stenotrophomonas maltophilia (▲) was detected significantly more commonly in AD patients, whilst Dietzia maris was detected significantly more commonly in normal controls Profile
Penders et al. (2007)55 AD Infants 957 (koala birth cohort) Cohort Feces Real time-PCR for bacterial DNA The presence of Escherichia coli (▲) was associated with a higher risk of developing eczema (OR, 1.87; 95% CI, 1.15–3.04). Infants colonised with Clostridium difficile (▲) were at higher risk of developing eczema (OR, 1.40; 95% CI, 1.02–1.91) and diagnosis of atopic dermatitis during the home visit (OR, 1.73; 95% CI, 1.08–2.78). Profile longitudinal
Wang et al. (2008)75 AD Infants 15 (case) 20 (control) Case-control Feces T-RFLP, Temporal temperature gradient gel electrophoresis (TTGE) analysis of 16S rRNA genes There is a reduced diversity (▼) in the early fecal microbiota of infants with atopic eczema during the first 18 months of life. Profile longitudinal
Fierer et al. (2008)40 Normal skin flora Adults 51 (healthy control) Observative Skin swab (hand) 16S rRNA gene pyrosequencing Propionibacteria (32%), Streptococcus (17%), Staphylococcus (8%), Corynebacterium (4%), Lactobacillus (3%) were the most abundant genera on palm surfaces. Profile
Grice et al. (2009)39 Normal skin flora Adults 10 (healthy control) Observative Skin swab and scraping 16S rRNA gene sequencing Propionibacteria species and staphylococci species predominated in sebaceous sites. Profile
Corynebacteria species predominated in moist sites, although staphylococci species were also represented. A mixed population of bacteria resided in dry sites, with a greater prevalence of β-Proteobacteria and Flavobacteriales.
Dominguez-Bello et al. (2010)76 Normal skin flora Mother & neonates 4 (vaginal delivery) 6 (cesarian section) Observative Mothers' skin, oral mucosa, vagina, neonates' skin, oral mucosa, nasopharyngeal aspirate 16S rRNA gene sequencing Vaginally delivered infants acquired bacterial communities resembling their own mother's vaginal microbiota, dominated by Lactobacillus, Prevotella, Sneathia spp., and C-section infants harbored bacterial communities similar to those found on the skin surface, dominated by Staphylococcus, Corynebacterium, Propionibacterium spp. Profile
De Filippo et al. (2010)51 Normal gut flora Children 15 (from burkina faso, africa) 15 (from florence, italy) Observative Feces 16S rRNA gene sequencing Burkina Faso children showed a significant enrichment in Bacteroidetes (▲) and depletion in Firmicutes (▼). Also, Enterobacteriaceae (Shigella and Escherichia) (▼) were significantly underrepresented in BF than in EU children. Profile
Capone et al. (2011)38 Normal skin flora Infants 31 (healthy infants) 5 (healthy mothers) Observative Skin swab 16S rRNA gene sequencing Composition of cutaneous microbial communities evolves over the first year of life, showing increasing diversity with age. Although early colonization is dominated by staphylococci, their significant decline contributes to increased population evenness by the end of the first year. Profile longitudinal
Human Microbiome Project Consortium (2012)16 Normal skin flora Aldults 242 (human microbiome project cohort) Cohort Oral cavity, skin, stool, vagina 16S rRNA gene sequencing Skin communities were dominated by one of Staphylococcus (Firmicutes), Propionibacterium, or Corynebacterium (Actinobacteria) with a continuum of oral organisms (Streptococcus) appearing in nares communities. Profile
Kong et al. (2012)46 AD Children 12 (case) 11 (placebo) Case-control Skin swab and scraping 16S rRNA gene sequencing In AD, the proportion of Staphylococcus sequences, particularly S. aureus (▲) was greater during disease flares than at baseline or post-treatment, and correlated with worsened disease severity. S. epidermidis (▲) also significantly increased during flares.
Increases in Streptococcus, Propionibacterium, and Corynebacterium species were observed following therapy.
Gosalbes et al. (2013)37 AD Infants 20 (term newborns) 2 (infants) 7 (pregnant women) Observative Feces, meconium 16S rRNA gene sequencing One of the types was less diverse, dominated by enteric bacteria (▲) and associated with a history of atopic eczema in the mother, whereas the second type was dominated by lactic acid bacteria (▲) and associated with respiratory problems in the infant. Profile
Seite et al. (2014)44 AD Children & adults 49 (case) Observative Skin swab 16S rRNA gene sequencing Overabundance of Staphylococcus species (▲) and a decrease in bacterial diversity were observed on affected skin. After 84-days of emollient treatment, increased overall diversity and a decrease in the Staphylococcus and Stenotrophomonas species were observed in treatment responders. Profile longitudinal

AD, atopic dermatitis; PCR, polymerase chain reaction; OR, odds ratio; CI, confidence interval; BF, Burkina Faso; EU, European Union; ▲, increase; ▼, decrease.

Notes

This research was supported by a fund by Research of Korea Centers for Disease Control and Prevention (2015ER660300).

References

1. NIAID-Sponsored Expert Panel. Boyce JA, Assa'ad A, Burks AW, Jones SM, Sampson HA, et al. Guidelines for the diagnosis and management of food allergy in the United States: report of the NIAID-sponsored expert panel. J Allergy Clin Immunol. 2010; 126:6 Suppl. S1–S58.
2. Du Toit G, Santos A, Roberts G, Fox AT, Smith P, Lack G. The diagnosis of IgE-mediated food allergy in childhood. Pediatr Allergy Immunol. 2009; 20:309–319.
crossref
3. Thomas CL, Fernández-Peñas P. The microbiome and atopic eczema: More than skin deep. Australas J Dermatol. 2016; 01. 28. [Epub]. DOI: 10.1111/ajd.12435.
crossref
4. Rona RJ, Keil T, Summers C, Gislason D, Zuidmeer L, Sodergren E, et al. The prevalence of food allergy: a meta-analysis. J Allergy Clin Immunol. 2007; 120:638–646.
crossref
5. Hong SJ, Ahn KM, Lee SY, Kim KE. The prevalences of asthma and allergic diseases in Korean children. Korean J Pediatr. 2008; 51:343–350.
crossref
6. Lee JH, Han KD, Kim KM, Park YG, Lee JY, Park YM. Prevalence of atopic dermatitis in Korean children based on data from the 2008-2011 Korean National Health and Nutrition Examination Survey. Allergy Asthma Immunol Res. 2016; 8:79–83.
crossref
7. Gupta R, Holdford D, Bilaver L, Dyer A, Holl JL, Meltzer D. The economic impact of childhood food allergy in the United States. JAMA Pediatr. 2013; 167:1026–1031.
crossref
8. Kim C, Park KY, Ahn S, Kim DH, Li K, Kim DW, et al. Economic Impact of Atopic Dermatitis in Korean Patients. Ann Dermatol. 2015; 27:298–305.
crossref
9. Lack G. Update on risk factors for food allergy. J Allergy Clin Immunol. 2012; 129:1187–1197.
crossref
10. Pyun BY. Natural history and risk factors of atopic dermatitis in children. Allergy Asthma Immunol Res. 2015; 7:101–105.
crossref
11. Allen KJ, Koplin JJ. Prospects for Prevention of Food Allergy. J Allergy Clin Immunol Pract. 2016; 4:215–220.
crossref
12. Molloy J, Allen K, Collier F, Tang ML, Ward AC, Vuillermin P. The potential link between gut microbiota and IgE-mediated food allergy in early life. Int J Environ Res Public Health. 2013; 10:7235–7256.
crossref
13. Baker BS. The role of microorganisms in atopic dermatitis. Clin Exp Immunol. 2006; 144:1–9.
crossref
14. Palmer CN, Irvine AD, Terron-Kwiatkowski A, Zhao Y, Liao H, Lee SP, et al. Common loss-of-function variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nat Genet. 2006; 38:441–446.
crossref
15. Strachan DP. Hay fever, hygiene, and household size. BMJ. 1989; 299:1259–1260.
crossref
16. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012; 486:207–214.
17. Kinross JM, Darzi AW, Nicholson JK. Gut microbiome-host interactions in health and disease. Genome Med. 2011; 3:14.
crossref
18. Choi S, Cho SH, Yi H. Human microbiome studies in Korea. Allergy Asthma Respir Dis. 2016; 4:311–320.
crossref
19. Kim BK, Rhee CK, Jung JY, Kang HR, Cho SH. Current status of microbiome research in asthma and chronic obstructive pulmonary disease. Allergy Asthma Respir Dis. 2016; 4:321–327.
crossref
20. NIH HMP Working Group. Peterson J, Garges S, Giovanni M, McInnes P, Wang L, et al. The NIH Human Microbiome Project. Genome Res. 2009; 19:2317–2323.
crossref
21. Noval Rivas M, Burton OT, Wise P, Zhang YQ, Hobson SA, Garcia Lloret M, et al. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol. 2013; 131:201–212.
crossref
22. Berni Canani R, Gilbert JA, Nagler CR. The role of the commensal microbiota in the regulation of tolerance to dietary allergens. Curr Opin Allergy Clin Immunol. 2015; 15:243–249.
crossref
23. Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, Mazmanian SK, et al. Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci U S A. 2014; 111:13145–13150.
crossref
24. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011; 331:337–341.
crossref
25. Rodriguez B, Prioult G, Hacini-Rachinel F, Moine D, Bruttin A, Ngom-Bru C, et al. Infant gut microbiota is protective against cow's milk allergy in mice despite immature ileal T-cell response. FEMS Microbiol Ecol. 2012; 79:192–202.
crossref
26. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013; 500:232–236.
crossref
27. Thompson-Chagoyan OC, Vieites JM, Maldonado J, Edwards C, Gil A. Changes in faecal microbiota of infants with cow's milk protein allergy--a Spanish prospective case-control 6-month follow-up study. Pediatr Allergy Immunol. 2010; 21(2 Pt 2):e394–e400.
28. Berni Canani R, Sangwan N, Stefka AT, Nocerino R, Paparo L, Aitoro R, et al. Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants. ISME J. 2016; 10:742–750.
crossref
29. Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT, et al. Infant gut microbiota and food sensitization: associations in the first year of life. Clin Exp Allergy. 2015; 45:632–643.
crossref
30. Hua X, Goedert JJ, Pu A, Yu G, Shi J. Allergy associations with the adult fecal microbiota: Analysis of the American Gut Project. EBioMedicine. 2015; 3:172–179.
crossref
31. Ling Z, Li Z, Liu X, Cheng Y, Luo Y, Tong X, et al. Altered fecal microbiota composition associated with food allergy in infants. Appl Environ Microbiol. 2014; 80:2546–2554.
crossref
32. Apostolou E, Pelto L, Kirjavainen PV, Isolauri E, Salminen SJ, Gibson GR. Differences in the gut bacterial flora of healthy and milk-hypersensitive adults, as measured by fluorescence in situ hybridization. FEMS Immunol Med Microbiol. 2001; 30:217–221.
crossref
33. Tang ML, Ponsonby AL, Orsini F, Tey D, Robinson M, Su EL, et al. Administration of a probiotic with peanut oral immunotherapy: a randomized trial. J Allergy Clin Immunol. 2015; 135:737–744.e8.
34. Fredricks DN. Microbial ecology of human skin in health and disease. J Investig Dermatol Symp Proc. 2001; 6:167–169.
crossref
35. Mshvildadze M, Neu J, Shuster J, Theriaque D, Li N, Mai V. Intestinal microbial ecology in premature infants assessed with non-culture-based techniques. J Pediatr. 2010; 156:20–25.
crossref
36. DiGiulio DB, Romero R, Amogan HP, Kusanovic JP, Bik EM, Gotsch F, et al. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS One. 2008; 3:e3056.
crossref
37. Gosalbes MJ, Llop S, Vallès Y, Moya A, Ballester F, Francino MP. Meconium microbiota types dominated by lactic acid or enteric bacteria are differentially associated with maternal eczema and respiratory problems in infants. Clin Exp Allergy. 2013; 43:198–211.
crossref
38. Capone KA, Dowd SE, Stamatas GN, Nikolovski J. Diversity of the human skin microbiome early in life. J Invest Dermatol. 2011; 131:2026–2032.
crossref
39. Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, et al. Topographical and temporal diversity of the human skin microbiome. Science. 2009; 324:1190–1192.
crossref
40. Fierer N, Hamady M, Lauber CL, Knight R. The influence of sex, handedness, and washing on the diversity of hand surface bacteria. Proc Natl Acad Sci U S A. 2008; 105:17994–17999.
crossref
41. Leyden JJ, Marples RR, Kligman AM. Staphylococcus aureus in the lesions of atopic dermatitis. Br J Dermatol. 1974; 90:525–530.
crossref
42. Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T, et al. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N Engl J Med. 2002; 347:1151–1160.
crossref
43. Gong JQ, Lin L, Lin T, Hao F, Zeng FQ, Bi ZG, et al. Skin colonization by Staphylococcus aureus in patients with eczema and atopic dermatitis and relevant combined topical therapy: a double-blind multicentre randomized controlled trial. Br J Dermatol. 2006; 155:680–687.
crossref
44. Seite S, Flores GE, Henley JB, Martin R, Zelenkova H, Aguilar L, et al. Microbiome of affected and unaffected skin of patients with atopic dermatitis before and after emollient treatment. J Drugs Dermatol. 2014; 13:1365–1372.
45. Huang JT, Abrams M, Tlougan B, Rademaker A, Paller AS. Treatment of Staphylococcus aureus colonization in atopic dermatitis decreases disease severity. Pediatrics. 2009; 123:e808–e814.
crossref
46. Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012; 22:850–859.
crossref
47. Seite S, Bieber T. Barrier function and microbiotic dysbiosis in atopic dermatitis. Clin Cosmet Investig Dermatol. 2015; 8:479–483.
crossref
48. Gallo RL, Nakatsuji T. Microbial symbiosis with the innate immune defense system of the skin. J Invest Dermatol. 2011; 131:1974–1980.
crossref
49. Dekio I, Sakamoto M, Hayashi H, Amagai M, Suematsu M, Benno Y. Characterization of skin microbiota in patients with atopic dermatitis and in normal subjects using 16S rRNA gene-based comprehensive analysis. J Med Microbiol. 2007; 56(Pt 12):1675–1683.
crossref
50. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006; 124:837–848.
crossref
51. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010; 107:14691–14696.
crossref
52. Watanabe S, Narisawa Y, Arase S, Okamatsu H, Ikenaga T, Tajiri Y, et al. Differences in fecal microflora between patients with atopic dermatitis and healthy control subjects. J Allergy Clin Immunol. 2003; 111:587–591.
crossref
53. Björkstén B, Sepp E, Julge K, Voor T, Mikelsaar M. Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol. 2001; 108:516–520.
crossref
54. Kalliomäki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin Immunol. 2001; 107:129–134.
crossref
55. Penders J, Thijs C, van den Brandt PA, Kummeling I, Snijders B, Stelma F, et al. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut. 2007; 56:661–667.
crossref
56. Ismail IH, Oppedisano F, Joseph SJ, Boyle RJ, Licciardi PV, Robins-Browne RM, et al. Reduced gut microbial diversity in early life is associated with later development of eczema but not atopy in high-risk infants. Pediatr Allergy Immunol. 2012; 23:674–681.
crossref
57. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol. 2012; 129:434–440. 440.e1–440.e2.
crossref
58. Nylund L, Satokari R, Nikkilä J, Rajilić-Stojanović M, Kalliomäki M, Isolauri E, et al. Microarray analysis reveals marked intestinal microbiota aberrancy in infants having eczema compared to healthy children in at-risk for atopic disease. BMC Microbiol. 2013; 13:12.
crossref
59. Ouwehand AC, Isolauri E, He F, Hashimoto H, Benno Y, Salminen S. Differences in Bifidobacterium flora composition in allergic and healthy infants. J Allergy Clin Immunol. 2001; 108:144–145.
crossref
60. Sanders ME. Probiotics: definition, sources, selection, and uses. Clin Infect Dis. 2008; 46:Suppl 2. S58–S61.
crossref
61. Kalliomäki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001; 357:1076–1079.
crossref
62. Kalliomäki M, Salminen S, Poussa T, Arvilommi H, Isolauri E. Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet. 2003; 361:1869–1871.
crossref
63. Isolauri E, Arvola T, Sütas Y, Moilanen E, Salminen S. Probiotics in the management of atopic eczema. Clin Exp Allergy. 2000; 30:1604–1610.
crossref
64. Wang IJ, Wang JY. Children with atopic dermatitis show clinical improvement after Lactobacillus exposure. Clin Exp Allergy. 2015; 45:779–787.
crossref
65. Drago L, De Vecchi E, Toscano M, Vassena C, Altomare G, Pigatto P. Treatment of atopic dermatitis eczema with a high concentration of Lactobacillus salivarius LS01 associated with an innovative gelling complex: a pilot study on adults. J Clin Gastroenterol. 2014; 48:Suppl 1. S47–S51.
66. Niccoli AA, Artesi AL, Candio F, Ceccarelli S, Cozzali R, Ferraro L, et al. Preliminary results on clinical effects of probiotic Lactobacillus salivarius LS01 in children affected by atopic dermatitis. J Clin Gastroenterol. 2014; 48:Suppl 1. S34–S36.
crossref
67. Boyle RJ, Bath-Hextall FJ, Leonardi-Bee J, Murrell DF, Tang ML. Probiotics for treating eczema. Cochrane Database Syst Rev. 2008; (4):CD006135.
crossref
68. Kim SO, Ah YM, Yu YM, Choi KH, Shin WG, Lee JY. Effects of probiotics for the treatment of atopic dermatitis: a meta-analysis of randomized controlled trials. Ann Allergy Asthma Immunol. 2014; 113:217–226.
crossref
69. Panduru M, Panduru NM, Sălăvăstru CM, Tiplica GS. Probiotics and primary prevention of atopic dermatitis: a meta-analysis of randomized controlled studies. J Eur Acad Dermatol Venereol. 2015; 29:232–242.
crossref
70. Gueniche A, Knaudt B, Schuck E, Volz T, Bastien P, Martin R, et al. Effects of nonpathogenic gram-negative bacterium Vitreoscilla filiformis lysate on atopic dermatitis: a prospective, randomized, double-blind, placebo-controlled clinical study. Br J Dermatol. 2008; 159:1357–1363.
crossref
71. Volz T, Skabytska Y, Guenova E, Chen KM, Frick JS, Kirschning CJ, et al. Nonpathogenic bacteria alleviating atopic dermatitis inflammation induce IL-10-producing dendritic cells and regulatory Tr1 cells. J Invest Dermatol. 2014; 134:96–104.
crossref
72. Mahe YF, Perez MJ, Tacheau C, Fanchon C, Martin R, Rousset F, et al. A new Vitreoscilla filiformis extract grown on spa water-enriched medium activates endogenous cutaneous antioxidant and antimicrobial defenses through a potential Toll-like receptor 2/protein kinase C, zeta transduction pathway. Clin Cosmet Investig Dermatol. 2013; 6:191–196.
73. Thompson-Chagoyan OC, Fallani M, Maldonado J, Vieites JM, Khanna S, Edwards C, et al. Faecal microbiota and short-chain fatty acid levels in faeces from infants with cow's milk protein allergy. Int Arch Allergy Immunol. 2011; 156:325–332.
crossref
74. Chen CC, Chen KJ, Kong MS, Chang HJ, Huang JL. Alterations in the gut microbiotas of children with food sensitization in early life. Pediatr Allergy Immunol. 2016; 27:254–262.
crossref
75. Wang M, Karlsson C, Olsson C, Adlerberth I, Wold AE, Strachan DP, et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol. 2008; 121:129–134.
crossref
76. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A. 2010; 107:11971–11975.
crossref
TOOLS
ORCID iDs

Dong In Suh
https://orcid.org/http://orcid.org/0000-0002-7817-8728

Similar articles