23

The Microbiome and Probiotics in Childhood Michael Harrison Hsieh, MD, PhD1

Stanford University, Stanford, California Semin Reprod Med 2014;32:23–27

Abstract

Keywords

► probiotics ► children ► microbiome

Address for correspondence Michael Harrison Hsieh, MD, PhD, Department of Urology, Stanford University School of Medicine, Stanford University, 300 Pasteur Drive, S-287, Stanford, CA 9305 (e-mail: [email protected]).

Infants, from the moment of birth, are colonized by large numbers of microbes. This colonization continues throughout childhood and from preliminary studies seems to be a highly dynamic process, even during the usual physiologic state we refer to as health. In this context, the persistence of bacterial and fungal species in and on the human body likely confers various benefits to the host. One specific approach to modulate such beneficial effects is the administration of probiotics, also known as beneficial microbes. Herein, we outline the highest level evidence in regard to the evolution of the microbiome during childhood and its manipulation by probiotics for genitourinary, enteric, and allergic and atopic disorders. Thus, probiotic approaches are promising alternatives and adjuvants to traditional vaccines and antibiotics. This may usher in a new age in which vaccine and antibiotic side effects and antibiotic resistance are minimal issues in the setting of maintaining children’s health and prevention of disease.

The Human Microbiome Project and other microbial genomics initiatives have primarily focused on the adult human microbiome. If the microbial landscape we call the microbiome is relatively unmapped in adults, by comparison the microbiome of children is almost completely unknown. Moreover, it is highly likely that the microbiome of children is particularly dynamic, given that the rapid growth and physiological changes which occur throughout childhood may influence the pediatric microbiome. In general, children also rebound more quickly from many illnesses compared with adults, another biological feature which may modulate the microbiome during childhood. Considering that probiotics, “live microorganisms which when administered in adequate amounts confer a health benefit on the host,”1 may mediate most of their effects through perturbations of the microbiome, it is probable that outcomes of microbial therapy will be heavily predicated on the age and developmental status of a given child. Herein, we review in detail the known literature regarding interactions among the microbiome, probiotics, and children’s health, with a goal of highlighting the highest quality findings.

The Known Pediatric Microbiome Before the quality of evidence for therapeutic microbiology (e.g., probiotics) in children can be weighed, we must first

Issue Theme The Microbiome and Reproduction; Guest Editors, James H. Segars, MD, and Kjersti M. Aagaard, MD, PhD

outline what is known regarding the microbial communities in and on children’s bodies in the baseline state we refer to as health. It has been estimated that 100 trillion microbes live within each individual human being.2,3 The continuous interactions among our bodies and these microbes partially determine whether individuals stay healthy or develop diseases such as infections and immune-mediated disorders. For instance, commensal bacteria in the gut can facilitate metabolism of bile acids and bile acid proteins, vitamin absorption and production, inhibition of pathogen overgrowth, and fermentation of dietary carbohydrates.4 Moreover, pathogens may have unexpected roles in diverse diseases including autoimmune disorders,5 cancer,6 inflammatory bowel disease (IBD),7 and chronic pain states.8,9 However, before the human microbiota modulates health and disease, natural birth, as the infant passes through the birth canal, is typically the first encounter with environmental microbes. These microbes include the endogenous microbiota of the mother’s vagina (except in cases of cesarean delivery) and distal enteric tract,10 but are modulated by maternal administration of antibiotics, such as treatment for Group B streptococcus to prevent transmission to the infant. Indeed, birth by cesarean section and neonatal antibiotic utilization delay infant gut colonization.11 Hospitalization, diet, and mode of feeding likely also influence the infant microbiota.12 Palmer et al

Copyright © 2014 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662.

DOI http://dx.doi.org/ 10.1055/s-0033-1361819. ISSN 1526-8004.

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

1 Department of Urology, Stanford University School of Medicine,

Microbiome and Probiotics in Childhood

Hsieh

reported that the composition and temporal patterns of enteric microbiota varied broadly among infants, with complex communities existing by 1 week of age and yet demonstrating stability for months at a time.11 Dizygotic twins featured parallel temporal patterns suggestive of environmental effects. At the end of the first year of life, each infant’s enteric microbiome achieves relative equilibrium and adult-like complexity. Interestingly, some have speculated that the composition of the intestinal microbiota is linked to brain development (reviewed by Douglas-Escobar et al13). This body of work, as well as other microbiome studies, has led to the concepts of eubiosis versus dysbiosis, normal versus abnormal microbiota, as key determinants of health and disease (reviewed by Buccigrossi et al14). The kinetics of bacterial colonization in the newborn gut are further complicated by passage of commensal bacteria (i.e., Bifidobacterium) from maternal blood to breast milk, and eventually the infant enteric tract.15 Bacterial components may translocate from mother to infant through maternal leukocytes, which in turn may train the developing infant immune system to appropriately respond to commensal and pathogenic bacteria. Certainly, more data are needed to explain the patterns of microbial colonization and their effects on the development of childhood immunity.

Probiotics for Pediatric Atopic/Allergic Disorders Probiotics may modulate host immune responses and thereby alleviate pediatric immune-mediated disorders. The immunomodulatory effects of commensal-derived probiotics have implications for the hygiene hypothesis. This hypothesis asserts that the increased incidence of chronic inflammatory diseases (e.g., asthma, allergic disorders, IBDs) is partly attributable to decreased early life exposure to commensal microbes and other organisms that have been tightly associated with humans throughout evolution.16 These organisms, including saprophytic mycobacteria and worms, may be viewed by the innate immune system as harmless commensals which induce immunoregulatory mechanisms. Indeed, contact with cows, straw, and consumption of unprocessed cow’s milk may provide protection against asthma (reviewed by Wlasiuk and Vercelli17). Many studies have been conducted to elucidate the role of probiotics in atopic diseases and allergies. These studies are too numerous to analyze individually herein, but one systematic review suggested that Lactobacillus rhamnosus GG may be effective in long-term prevention of development of atopic dermatitis.18 Another meta-analysis reported that probiotics reduced the incidence of atopic dermatitis regardless of whether use was during pregnancy, early life, or taken by either mother and/or child (relative risk [RR], 0.79; 95% confidence interval [CI], 0.71–0.88]).19 Interestingly, a third meta-analysis found that Lactobacillus-only probiotics given during pregnancy reduced risk of atopic eczema in children, but multistrain probiotics did not, even if they contained lactobacilli.20 Thus, several systematic reviews and metaanalyses suggest that probiotics, and perhaps lactobacilli in particular, may reduce risk of atopic dermatitis. Seminars in Reproductive Medicine

Vol. 32

No. 1/2014

Probiotics for Enteric Health and Disease The use of probiotics in infant formula is another unresolved application of probiotics. Braegger et al have declared that although probiotic-supplemented formula does not appear to adversely affect infant growth or result in apparent ill effects, they caution that there is still insufficient data to recommend the widespread use of probiotic-supplemented formula.21 Mugambi et al likewise agree that weight gain is unaltered by use of probiotic-supplemented formula, but that the number of stools per day increased significantly with the use of supplemented formula.22 In addition, probiotic-supplemented formula did not decrease the incidence of infant gastroesophageal reflux, crying, restlessness, vomiting, or diarrhea.23 In premature infants, supplementation with Bifidobacterium lactis had no effect on the risk of necrotizing enterocolitis stage  or 2 (RR, 0.53; 95% CI, 0.16–1.83), risk of sepsis (RR, 0.6; 95% CI, 0.07–5.2), or need for antibiotics (RR, 0.67; 95% CI, 0.28–1.62). A lack of power of these studies, however, does not allow for a clear statement regarding effects on risk of necrotizing enterocolitis. Interestingly, B. lactis supplementation did have some effects on anthropometric measures. No adverse events associated with B. lactis supplementation were observed.24 In a broader meta-analysis encompassing numerous probiotic supplements, formula supplementation was associated with a decreased risk of necrotizing enterocolitis in preterm very low-birth-weight infants (RR, 0.33; 95% CI, 0.24–0.46). Risk of death was also reduced in the probiotic supplementation group (RR, 0.56; 95% CI, 0.43–0.73). However, there was no difference in the risk of sepsis between the probiotic and placebo groups (RR, 0.90; 95% CI, 0.71–1.15).25 Very similar findings were reported by a Cochrane Review by Alfaleh et al,26 and Downard et al27 in another Cochrane Review likewise advocated the use of prophylactic probiotics in preterm infants (< 2,500 g) to reduce the incidence of necrotizing enterocolitis. Thus, probiotic supplementation of formula in general may be useful for prevention of necrotizing enterocolitis and reducing risk of death in premature infants. Probiotics have also been tested in the setting of other enteric diseases, namely, Helicobacter pylori infection. One trial of children infected with H. pylori demonstrated that L. casei DN-114 001 supplementation of triple therapy (omeprazole, amoxicillin, and clarithromycin) yielded greater H. pylori clearance rates.28 One meta-analysis showed that Saccharomyces boulardii supplementation of triple therapy enhanced the eradication rate (RR, 11.13; 95% CI, 1.05–1.21) and reduced the risk of therapy-related adverse effects (RR, 0.46; 95% CI, 0.3–0.7), especially diarrhea (RR, 0.47; 95% CI, 0.32–0.69).29 Thus, both Lactobacillus and S. boulardii supplementation of triple therapy may augment its anti-H. pylori effects. Another gastrointestinal disease that has been treated with probiotics is antibiotic-associated diarrhea. Johnston et al concluded, in a Cochrane Review, that despite heterogeneous probiotic strains, doses, duration, and study quality, there may be a protective effect of probiotics against the

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

24

development of antibiotic-associated diarrhea, but that the quality of the positive evidence was poor.30 A specific form of antibiotic-associated diarrhea with relevance to probiotics and children is C. difficile-associated infections. A systematic review and meta-analysis of the literature demonstrated that probiotics reduced the incidence of C. difficile-associated diarrhea by 66% (pooled RR, 0.34 [95% CI, 0.24–0.49]). Among probiotic-treated patients, 9.3% experienced adverse events, compared with 12.6% of control patients (RR, 0.82 [95% CI, 0.65–1.05]).31 However, in a subsequent systematic review and meta-analysis of the effects of probiotics on the risk of C. difficile-associated, antibiotic-induced diarrhea in adults and children, there was no difference in incidence rates of infection among the probiotics, placebo, or no treatment control groups (RR, 0.89; 95% CI, 0.64–1.24). A pooled analysis of adverse events indicated that probiotics reduce the risk of adverse events by 20% (RR, 0.80; 95% CI, 0.68–0.95).32 Thus, the “jury is still out” for the efficacy of probiotics in C. difficile-associated, antibiotic-induced diarrhea in children. In other forms of diarrhea, namely infectious, S. boulardii has been specifically noted as being effective.33 For example, S. boulardii reduced the duration of diarrhea by 1 day and hospitalization by 20 hours. Lactobacillus rhamnosus GG also seems to reduce rates of diarrhea (RR, 0.37; 95% CI, 0.23–0.59) and symptomatic rotavirus gastroenteritis (RR, 0.49; 95% CI, 0.28–0.86). However, there was no significant difference between the probiotic and placebo groups in duration of hospitalization or diarrhea. Lactobacillus rhamnosus GG was not associated with harm in any of the trials.34 Ostensibly noninfectious pediatric gastrointestinal disorders, specifically abdominal pain, have also been explored as therapeutic targets of probiotics. One meta-analysis noted that L. rhamnosus GG supplementation was correlated with a significantly higher rate of treatment responses (e.g., decrease in pain intensity or no pain) in the overall population with abdominal pain-related functional enteric disorders (RR, 1.31; 95% CI, 1.08–1.59 and 4–22) and in the irritable bowel syndrome subgroup (RR, 1.70; 95% CI, 1.27–2.27 and 3–8).35 Thus, Lactobacillus-based probiotics may have a beneficial role in these enteric diseases. In contrast, the utility of probiotics in pediatric IBD (i.e., Crohn disease or ulcerative colitis) is unclear at best. A metaanalysis indicated that IBD patients receiving lactobacilli featured essentially equivalent rates of clinical or endoscopic relapse, even among children.36 This was true whether patients received L. rhamnosus strain GG or Lactobacillus johnsonii.36

The Microbiome and Pediatric Obesity: A Potential Role for Probiotics The enteric microbiota may modulate the host not only locally within the gut; commensal microbes may also confer systemic effects and profoundly influence host metabolism. For example, one article suggested that the sizes of gut populations of Bacteroides and Firmicutes bacteria (including their ratios) may be associated with pediatric obesity.37 In another

Hsieh

publication, the relative size of Enterobacteriaceae, Desulfovibrio, and Akkermansia muciniphila-like bacteria was also related to risk of obesity in children.38 These differences in the gut microbiome of lean and obese children suggest that probiotic manipulation could influence metabolism. However, Kaplan and Walker have correctly pointed out that establishing causality between the composition of enteric microbial communities and obesity is difficult.39 Conversely, the microbiome may be important in states of chronic starvation in children such as kwashiorkor. Smith et al studied Malawian twin pairs discordant for nutritional state (i.e., one twin remain nourished, whereas the other malnourished).40 Both children in discordant twin pairs were given a peanut-based, ready-to-use therapeutic food (RUTF). Metagenomic studies showed that RUTF induced a transient maturation of metabolic functions in kwashiorkor-associated enteric microbiomes that regressed upon cessation of RUTF. Moreover, transplantation of feces from kwashiorkor patients on a Malawian diet into gnotobiotic mice resulted in weight loss and perturbations in metabolism that were only transiently rescued by RUTF. Thus, the gut microbiome may be a key causal parameter in pediatric starvation states such as kwashiorkor.40

Probiotics for Urogenital Health and Disease In the 1970s, workers ascertained that most uropathogens in girls originate in the gut, traverse the perineum to the vagina, and then infect the bladder. Some investigators have examined the microbiology of indigenous lactobacilli found in high numbers in the vaginas of women with no history of urinary tract infections (UTI), and how the absence of these commensal microbes may be associated with susceptibility to recurrent UTI.41 As a consequence of this observation, other workers have postulated that it may be possible to favorably manipulate the vaginal microbiome through orally administered probiotics. In one clinical trial, 585 premature infants (< 33 weeks or < 1,500 g) were fed milk supplemented with 6  109 colony-forming unit (CFU) of L. rhamnosus GG or placebo milk once per day from the initial feed until discharge (mean 48 days).42 The frequency of UTI episodes was diminished (3.4 vs. 5.8%), but the difference was not statistically significant. A randomized clinical trial compared oral L. acidophilus ATCC 4356 to trimethoprim/sulfamethoxazole as prophylaxis for UTI in 120 children with vesicoureteral reflux.43 The two treatment arms did not differ significantly with regard to UTI prevention with incidences of 18.3% in the probiotic arm and 21.6% in the antibiotic arm. However, these results indicate that probiotics could be used as alternatives to antibiotics with a possible reduction of complications due to antibiotics. A more complex question beyond the scope of this review is whether and in what settings antibiotic prophylaxis is efficacious for children with reflux. Regardless, more studies are needed to confirm these findings. In summary, there is little data on probiotic applications to prevent pediatric UTI. An intriguing but unexplored Seminars in Reproductive Medicine

Vol. 32

No. 1/2014

25

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Microbiome and Probiotics in Childhood

Microbiome and Probiotics in Childhood

Hsieh

possibility is combining antibiotics with probiotics. If combinations of antibiotics and probiotics are efficacious, these agents may help prevent or cure UTI with less antibiotic resistance and fewer antibiotic-associated side effects.

Probiotics-Induced Sepsis Although probiotics seem to be promising approaches for prevention or treatment of a variety of pediatric diseases and are mostly well-tolerated, several reports of probioticsassociated bacteremia and sepsis in children have been published.44,45 Some of these studies included infants who are susceptible to infections due to immature immune systems and underdeveloped mucosal barriers. Caution with regard to selection of probiotics strains (such as focusing on commensalderived probiotics), and clinical study design, particularly in infants, is warranted. Given the large numbers of commercially available probiotic products that are poorly or mislabeled,46–56 we advise that patients and their health care providers only consider products which document not only specific bacterial species but full strain names, CFU, nonprobiotic ingredients, manufacturing standards, and high quality, peer-reviewed scientific studies of the probiotic strains in question.

1 Guidelines for the Evaluation of Probiotics in Food. Geneva,

2 3

4

5 6

7

8

9 10

11

Summary Advances in the science of probiotics for children in the 21st century will be accelerated by pediatric microbiome research and functional genomics of beneficial microbes in children. These commensal microbes or “Old Friends” mediate development of robust immune responses, digestion, absorption of vitamins, and suppression of pathogens. Two major pipelines will fuel the development of new probiotics in pediatric medicine. First, the delineation of genes responsible for crucial probiotic functions will facilitate derivation of genetically modified organisms, also known as “designer strains,” that will represent optimized, engineered alternatives to natural probiotic strains for specific pediatric applications. Second, candidate probiotic strains isolated from natural sources (human, animal, or food) can be compared systematically by functional genomics and systems biology. The best natural probiotics can then be selected for specific pediatric uses. Many more clinical and laboratory investigations must be performed to elucidate mechanisms of probiosis and matching of specific probiotics with particular disease phenotypes in children. In summary, humans have used beneficial bacteria via the diet throughout human history. Investigator-initiated research and “big science,” as exemplified by the Human Microbiome Project, will drive accelerated development of probiotics for the prevention and treatment of many pediatric disorders.

Acknowledgment M.H.H. has research support from the National Institutes of Health (NIH) (NIDDK K08 DK087895).

Seminars in Reproductive Medicine

References

Vol. 32

No. 1/2014

12

13 14 15

16

17

18

19

20

21

22

Switzerland: the Food and Agriculture Organization of the United Nations and World Health Organization; 2002:1–11 Tierno PM. The Secret Life of Germs. New York: Atria; 2001 Paustian T. The normal flora of humans. In: Paustian T, ed. Microbiology and Bacteriology: The World of Microbes. Madison, WI: University of Wisconsin-Madison; 2006 Rolfe RD. Interactions among microorganisms of the indigenous intestinal flora and their influence on the host. Rev Infect Dis 1984; 6(Suppl 1):S73–S79 Wucherpfennig KW. Structural basis of molecular mimicry. J Autoimmun 2001;16(3):293–302 Nackerdien ZE. Perspectives on microbes as oncogenic infectious agents and implications for breast cancer. Med Hypotheses 2008; 71(2):302–306 Giaffer MH, Holdsworth CD, Duerden BI. The assessment of faecal flora in patients with inflammatory bowel disease by a simplified bacteriological technique. J Med Microbiol 1991; 35(4):238–243 Barbara G, Stanghellini V, Brandi G, et al. Interactions between commensal bacteria and gut sensorimotor function in health and disease. Am J Gastroenterol 2005;100(11):2560–2568 Quigley EM. Germs, gas and the gut; the evolving role of the enteric flora in IBS. Am J Gastroenterol 2006;101(2):334–335 Vaishampayan PA, Kuehl JV, Froula JL, Morgan JL, Ochman H, Francino MP. Comparative metagenomics and population dynamics of the gut microbiota in mother and infant. Genome Biol Evol 2010;2:53–66 Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Development of the human infant intestinal microbiota. PLoS Biol 2007; 5(7):e177 Murgas Torrazza R, Neu J. The developing intestinal microbiome and its relationship to health and disease in the neonate. J Perinatol 2011;31(Suppl 1):S29–S34 Douglas-Escobar M, Elliott E, Neu J. Effect of intestinal microbial ecology on the developing brain. JAMA Pediatr 2013;167(4):374–379 Buccigrossi V, Nicastro E, Guarino A. Functions of intestinal microflora in children. Curr Opin Gastroenterol 2013;29(1):31–38 Perez PF, Doré J, Leclerc M, et al. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics 2007;119(3):e724–e732 Rook GA. The hygiene hypothesis and the increasing prevalence of chronic inflammatory disorders. Trans R Soc Trop Med Hyg 2007; 101(11):1072–1074 Wlasiuk G, Vercelli D. The farm effect, or: when, what and how a farming environment protects from asthma and allergic disease. Curr Opin Allergy Clin Immunol 2012;12(5):461–466 Foolad N, Brezinski EA, Chase EP, Armstrong AW. Effect of nutrient supplementation on atopic dermatitis in children: a systematic review of probiotics, prebiotics, formula, and fatty acids. JAMA Dermatol 2013;149(3):350–355 Pelucchi C, Chatenoud L, Turati F, et al. Probiotics supplementation during pregnancy or infancy for the prevention of atopic dermatitis: a meta-analysis. Epidemiology 2012;23(3):402–414 Doege K, Grajecki D, Zyriax B-C, Detinkina E, Zu Eulenburg C, Buhling KJ. Impact of maternal supplementation with probiotics during pregnancy on atopic eczema in childhood—a meta-analysis. Br J Nutr 2012;107(1):1–6 Braegger C, Chmielewska A, Decsi T, et al; ESPGHAN Committee on Nutrition. Supplementation of infant formula with probiotics and/ or prebiotics: a systematic review and comment by the ESPGHAN committee on nutrition. J Pediatr Gastroenterol Nutr 2011;52(2): 238–250 Mugambi MN, Musekiwa A, Lombard M, Young T, Blaauw R. Probiotics, prebiotics infant formula use in preterm or low birth weight infants: a systematic review. Nutr J 2012;11:58

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

26

23 Mugambi MN, Musekiwa A, Lombard M, Young T, Blaauw R.

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

Synbiotics, probiotics or prebiotics in infant formula for full term infants: a systematic review. Nutr J 2012;11:81 Szajewska H, Guandalini S, Morelli L, Van Goudoever JB, Walker A. Effect of Bifidobacterium animalis subsp lactis supplementation in preterm infants: a systematic review of randomized controlled trials. J Pediatr Gastroenterol Nutr 2010;51(2):203–209 Wang Q, Dong J, Zhu Y. Probiotic supplement reduces risk of necrotizing enterocolitis and mortality in preterm very lowbirth-weight infants: an updated meta-analysis of 20 randomized, controlled trials. J Pediatr Surg 2012;47(1):241–248 Alfaleh K, Anabrees J, Bassler D, Al-Kharfi T. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev 2011;(3):CD005496 Downard CD, Renaud E, St Peter SD, et al; 2012 American Pediatric Surgical Association Outcomes Clinical Trials Committee. Treatment of necrotizing enterocolitis: an American Pediatric Surgical Association Outcomes and Clinical Trials Committee systematic review. J Pediatr Surg 2012;47(11):2111–2122 Sýkora J, Valecková K, Amlerová J, et al. Effects of a specially designed fermented milk product containing probiotic Lactobacillus casei DN-114 001 and the eradication of H. pylori in children: a prospective randomized double-blind study. J Clin Gastroenterol 2005;39(8):692–698 Szajewska H, Horvath A, Piwowarczyk A. Meta-analysis: the effects of Saccharomyces boulardii supplementation on Helicobacter pylori eradication rates and side effects during treatment. Aliment Pharmacol Ther 2010;32(9):1069–1079 Johnston BC, Goldenberg JZ, Vandvik PO, Sun X, Guyatt GH. Probiotics for the prevention of pediatric antibiotic-associated diarrhea. Cochrane Database Syst Rev 2011;(11):CD004827 Johnston BC, Ma SSY, Goldenberg JZ, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea: a systematic review and meta-analysis. Ann Intern Med 2012;157(12): 878–888 Goldenberg JZ, Ma SS, Saxton JD, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev 2013;5(CD006095):CD006095 Dinleyici EC, Eren M, Ozen M, Yargic ZA, Vandenplas Y. Effectiveness and safety of Saccharomyces boulardii for acute infectious diarrhea. Expert Opin Biol Ther 2012;12(4):395–410 Szajewska H, Wanke M, Patro B. Meta-analysis: the effects of Lactobacillus rhamnosus GG supplementation for the prevention of healthcare-associated diarrhoea in children. Aliment Pharmacol Ther 2011;34(9):1079–1087 Horvath A, Dziechciarz P, Szajewska H. Meta-analysis: Lactobacillus rhamnosus GG for abdominal pain-related functional gastrointestinal disorders in childhood. Aliment Pharmacol Ther 2011; 33(12):1302–1310 Shen J, Ran HZ, Yin MH, Zhou TX, Xiao DS. Meta-analysis: the effect and adverse events of Lactobacilli versus placebo in maintenance therapy for Crohn disease. Intern Med J 2009;39(2):103–109 Xu P, Li M, Zhang J, Zhang T. Correlation of intestinal microbiota with overweight and obesity in Kazakh school children. BMC Microbiol 2012;12:283 Karlsson CLJ, Onnerfält J, Xu J, Molin G, Ahrné S, Thorngren-Jerneck K. The microbiota of the gut in preschool children with normal and

39 40

41 42

43

44

45

46

47

48 49

50

51

52

53 54 55

56

Hsieh

excessive body weight. Obesity (Silver Spring) 2012;20(11): 2257–2261 Kaplan JL, Walker WA. Early gut colonization and subsequent obesity risk. Curr Opin Clin Nutr Metab Care 2012;15(3):278–284 Smith MI, Yatsunenko T, Manary MJ, et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science 2013; 339(6119):548–554 Bruce AW, Chadwick P, Hassan A, VanCott GF. Recurrent urethritis in women. Can Med Assoc J 1973;108(8):973–976 Dani C, Biadaioli R, Bertini G, Martelli E, Rubaltelli FF. Probiotics feeding in prevention of urinary tract infection, bacterial sepsis and necrotizing enterocolitis in preterm infants. A prospective double-blind study. Biol Neonate 2002;82(2):103–108 Lee SJ, Shim YH, Cho SJ, Lee JW. Probiotics prophylaxis in children with persistent primary vesicoureteral reflux. Pediatr Nephrol 2007;22(9):1315–1320 Salminen MK, Tynkkynen S, Rautelin H, et al. Lactobacillus bacteremia during a rapid increase in probiotic use of Lactobacillus rhamnosus GG in Finland. Clin Infect Dis 2002;35(10):1155–1160 Land MH, Rouster-Stevens K, Woods CR, Cannon ML, Cnota J, Shetty AK. Lactobacillus sepsis associated with probiotic therapy. Pediatrics 2005;115(1):178–181 Coeuret V, Gueguen M, Vernoux JP. Numbers and strains of lactobacilli in some probiotic products. Int J Food Microbiol 2004;97(2):147–156 Drisko J, Bischoff B, Giles C, Adelson M, Rao RV, McCallum R. Evaluation of five probiotic products for label claims by DNA extraction and polymerase chain reaction analysis. Dig Dis Sci 2005;50(6):1113–1117 Elliot E, Teversham K. An evaluation of nine probiotics available in South Africa, August 2003. S Afr Med J 2004;94(2):121–124 Hamilton-Miller JM, Shah S, Winkler JT. Public health issues arising from microbiological and labelling quality of foods and supplements containing probiotic microorganisms. Public Health Nutr 1999;2(2):223–229 Huys G, Vancanneyt M, D’Haene K, Vankerckhoven V, Goossens H, Swings J. Accuracy of species identity of commercial bacterial cultures intended for probiotic or nutritional use. Res Microbiol 2006;157(9):803–810 Szajewska H, Fordymacka A, Bardowski J, Górecki RK, Mrukowicz JZ, Banaszkiewicz A. Microbiological and genetic analysis of probiotic products licensed for medicinal purposes. Med Sci Monit 2004;10(9):BR346–BR350 Temmerman R, Scheirlinck I, Huys G, Swings J. Culture-independent analysis of probiotic products by denaturing gradient gel electrophoresis. Appl Environ Microbiol 2003;69(1):220–226 Weese JS. Microbiologic evaluation of commercial probiotics. J Am Vet Med Assoc 2002;220(6):794–797 Weese JS. Evaluation of deficiencies in labeling of commercial probiotics. Can Vet J 2003;44(12):982–983 Drago L, De Vecchi E, Nicola L, Colombo A, Gismondo MR. Microbiological evaluation of commercial probiotic products available in Italy. J Chemother 2004;16(5):463–467 Masco L, Huys G, De Brandt E, Temmerman R, Swings J. Culturedependent and culture-independent qualitative analysis of probiotic products claimed to contain bifidobacteria. Int J Food Microbiol 2005;102(2):221–230

Seminars in Reproductive Medicine

Vol. 32

No. 1/2014

27

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Microbiome and Probiotics in Childhood

Copyright of Seminars in Reproductive Medicine is the property of Thieme Medical Publishing Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

The microbiome and probiotics in childhood.

Infants, from the moment of birth, are colonized by large numbers of microbes. This colonization continues throughout childhood and from preliminary s...
138KB Sizes 0 Downloads 0 Views