You are on page 1of 12

FOCUS

The production of a variety of neuroactive signaling molecules by


bacterial components of the microbiome provides additional evidence
that the gut microbiome may generate signals with remote effects in
different organ systems, including the central nervous system

Ann Nutr Metab 2013;63(suppl 2):42–52


The Human Microbiome and Probiotics: Implications for Pediatrics
by James Versalovic

Key insights
Knowledge of the composition and function of the human Gastrointestinal
microbiome at multiple body sites including the gut, skin and
airways contributes to our understanding of the mechanisms
of probiosis. In turn, an enhanced understanding of the effects
Urogenital
of probiotics on the microbiome should facilitate selection of Oral
PC2 (4.4%)

optimal probiotic strains for the treatment of specific diseases.


Skin
Current knowledge
The human microbiome is composed of bacteria, viruses (in-
cluding bacteriophages), fungi, archaea and protozoa. Each
body site has its own distinct microbiome, with a unique mi-
Nasal
crobial composition that presumably reflects the differences in
tissue structure and function. For decades, it has been widely PC1 (13%)

accepted that the ingestion of probiotic strains has beneficial


Each body site has a specific microbiome. This coordinates plot displays
effects. The work of the Human Microbiome Project has pro- variation in terms of bacterial composition at different body sites in hu-
vided a wealth of data on the taxonomic profiles of over 5,000 mans; depicted here are the oral (darkest grey), gastrointestinal (light
bacterial strains from 18 different body sites. A major challenge grey), vaginal (dark grey), nasal (black) and skin (grey) microbiomes. For
is to quantify the relative abundance of the different strains in further details, please refer to the article. Adapted from Huttenhower et
health and disease, before we can harness the full potential of al. [Nature 2012;486:207–214].
probiotics.
tion of probiotics for NEC has been extended to the treatment
Practical implications of other intestinal disorders and provides a paradigm for the
Knowledge of the intestinal microbiome provides an opportu- therapy of other conditions such as asthma.
nity for understanding the roles of the microbial populations in
other parts of the body. The compositional differences between Recommended reading
the gut microbiomes of infants who develop necrotizing en- McNulty NP, Yatsunenko T, Hsiao A, Faith JJ, Muegge BD, Good-
terocolitis (NEC) versus healthy infants underscore the need to man AL, Henrissat B, Oozeer R, Cools-Portier S, Gobert G, Chervaux
tip the balance in favor of beneficial strains. Recent studies have C, Knights D, Lozupone CA, Knight R, Duncan AE, Bain JR, Muehl-
demonstrated that probiotics or symbiotics can have a direct bauer MJ, Newgard CB, Heath AC, Gordon JI: The impact of a con-
impact on gut microbial composition and profoundly affect the sortium of fermented milk strains on the gut microbiome of gno-
clinical outcomes for infants with NEC. The successful applica- tobiotic mice and monozygotic twins. Sci Transl Med 2011;3:106ra.
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

© 2013 S. Karger AG, Basel


Downloaded by:

E-Mail karger@karger.com
www.karger.com/anm
Ann Nutr Metab 2013;63(suppl 2):42–52 Published online: November 8, 2013
DOI: 10.1159/000354899

The Human Microbiome and Probiotics:


Implications for Pediatrics
James Versalovic
Department of Pathology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Tex., USA

Key Messages knowledge about the composition of the microbiome is pro-


• The composition and function of the microbiome can gressing rapidly, many gaps exist about the functional ca-
pacity and metabolic machinery of the human microbiome.
be changed by probiotics.
• The composition of the human microbiome at each Based on a limited amount of data, probiotics appear capa-
ble of altering the composition and function of the microbi-
body site is distinct, and different probiotics are likely
needed for diseases at different body sites. ome. Probiotics may be part of dietary strategies that com-
• Microbial deficiencies may be treated by probiotics as bine ways to enhance microbiome function with nutrients
that may be converted to active compounds promoting hu-
a strategy for microbial supplementation and
promotion of microbial diversity. man health. Probiotics have yielded beneficial effects in nu-
• Probiotics may change the ability of the microbiome merous studies in the context of different diseases in pedi-
atric gastroenterology. These disease states include necro-
to produce nutrients and bioactive compounds by de
novo biosynthesis or by luminal conversion. tizing enterocolitis, antibiotic-associated diarrhea and colitis,
acute gastroenteritis and irritable bowel syndrome. In the
skin and airways, it is unclear if probiotics can affect the func-
tion of the microbiome to reduce the impact of diseases such
Key Words as asthma and atopic dermatitis. An enhanced understand-
Human microbiome · Probiotics · Bacteria · Antibiotics · ing of the effects of probiotics on the microbiome should
Microbes · Metagenomics · Beneficial microbes · Dysbiosis facilitate selection of optimal probiotic strains for specific
diseases in the future. © 2013 S. Karger AG, Basel

Abstract
Steady advances in our knowledge of the composition and
function of the human microbiome at multiple body sites Pediatrics, the Microbiome and Probiotics
including the gut, skin and airways will likely contribute to The practice of pediatrics, as in other medical special-
our understanding of mechanisms of probiotic action by ties, has viewed microorganisms with a defensive posture
beneficial microbes. Microbe:microbe and microbe:human and considered each bacterium, fungus or virus as a po-
interactions are important considerations as we select pro- tential infectious agent. The prevailing ‘infectious diseas-
biotics for pediatric patients in the future. Although our es/antimicrobial’ strategic world view in medicine be-
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

© 2013 S. Karger AG, Basel James Versalovic, MD, PhD


0250–6807/13/0636–0042$38.00/0 Department of Pathology, Texas Children’s Hospital
1102 Bates Street, FC Suite 830
Downloaded by:

E-Mail karger@karger.com
Houston, TX 77030 (USA)
www.karger.com/anm
E-Mail jamesv @ bcm.edu
came a predominant view since the first 3 decades of the tered in adequate amounts confer a health benefit on the
twentieth century. As infectious agents and infectious host’ [8].
diseases were being characterized a century ago, the be- The emergence of investigations concerning the na-
ginning of antimicrobial agent discovery and consider- ture and mechanisms of probiosis during the 1990s and
ation of antibiotics as novel treatments began to take the rapid coalescence of the human microbiome research
shape. In fact, phage therapy as an antimicrobial strategy community globally since 2005 [9] provided the founda-
in medicine was being considered during the second de- tion for the current era in metagenomics (the genomic
cade of the 1900s (reviewed by Pirisi [1] and Keen [2]), analysis of microorganisms by direct extraction and clon-
and arsphenamine (Salvarsan) and its less toxic derivative ing of DNA from an assemblage of microorganisms). Hu-
(Neosalvarsan), discovered by Ehrlich and Hata in 1910, man microbiology includes canonical and opportunistic
were being applied to treat infectious agents, but this field
different infections. In 1928
(reprinted by Fleming [3]), Sir
Human-associated bacterial species has rapidly expanded to em-
brace the many commensal
Alexander Fleming’s discov- comprise the vast majority and beneficial microbes that
eries led to the identification of the human microbiome in may contribute to human
of antibacterial compounds terms of microbial DNA content health and disease prevention.
produced by specific fungi The specialty of pediatrics has
and laid the foundation for
and cell count. been swept into this new era
the antimicrobial era. This of human microbiology and
prevailing world view in medicine dominated the land- medicine as a result of numerous publications describing
scape of pediatrics until the first decade of this century, the composition of the microbiome in children and dif-
when the attitudes towards commensal and beneficial mi- ferences in the microbiome associated with diseases of
crobes began to change profoundly. childhood [10]. Alterations in microbial composition as-
Beneficial microbes and more specifically probiotics sociated with human diseases have been described as ex-
were described initially by Nobel laureate Elie Metch- amples of dysbiosis. Dysbiosis refers to differences in mi-
nikoff [4] in 1907/1908, when he described the potential crobial populations that may reflect an abnormal ecolog-
benefits of consumption of large quantities of microbes ical state contributing to pathology or the excess of
to improve longevity and human health. Unfortunately, pathogenic mechanisms within the human microbiome
this viewpoint was effectively subordinated to the view [see Chan et al. in this issue]. Functional components of
that microbes must be considered as potentially infec- the microbiome may be studied by determination of
tious agents, and most attention in the medical profes- DNA sequences or genes present in the microbiome, but
sion including pediatrics turned to vaccine development this information only reveals the metabolic capacity.
and antibiotic production. The term ‘probiotic’ was first RNA sequencing and metabolomics studies are necessary
credited to Lilly and Stillwell [5] who proposed this term to determine which microbial genes are expressed and
in the context of microbes producing substances that which metabolites may affect disease susceptibilities in
promoted the growth of other microorganisms. Parker children.
[6] was the first to use the term ‘probiotic’ to describe The human microbiome is composed of bacteria, vi-
microorganisms (and substances) that have beneficial ruses (including bacteriophages), fungi, archaea and pro-
effects on a host animal. Outside of the dairy and fer- tozoa in declining order. Human-associated bacterial
mented food industry, the probiotic concept remained species comprise the vast majority of the human micro-
largely dormant until the late 1980s. The British Profes- biome in terms of microbial DNA content and cell count.
sor R. Fuller [7] described the modern probiotic concept In fact, in one recent study, more than 99% of mapped
in a landmark review published in 1989 and proposed DNA sequencing reads in healthy adults were bacterial
the importance of microbial viability to probiotic func- sequences [11]. Human-associated bacterial communi-
tion. A formal definition of probiotics was formulated in ties are composed of four dominant phyla (Actinobacte-
2001 by the advisory body of the Food and Agriculture ria, Bacteroidetes, Firmicutes and Proteobacteria) and a
Organization (FAO) and the World Health Organiza- number of minority phyla [12]. A recent study of the bio-
tion (WHO), and this definition has been widely utilized geography of the human microbiome identified 30 phyla
during the past 12 years. This definition states that pro- in 18 different body sites in neonates and adults [13], and
biotics are ‘live microorganisms which when adminis- reports of cultured and uncultured bacteria estimates ap-
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

The Human Microbiome and Probiotics Ann Nutr Metab 2013;63(suppl 2):42–52 43
DOI: 10.1159/000354899
Downloaded by:
rial genera [16]. The initial comprehensive summary of
the Human Microbiome Project included 242 individuals
with greater than 5,000 bacterial taxonomic profiles from
Gastrointestinal
18 body sites in healthy adults [11, 12]. The Firmicutes
and Bacteroidetes were the dominant phyla in the gastro-
intestinal tract, and Lactobacillus was a minority Fir-
micute genus in these individuals. The phylum Actino-
Urogenital
Oral
bacteria includes the genus Bifidobacterium which has
PC2 (4.4%)

been underrepresented in human microbiome studies


due to technical challenges in DNA amplification/detec-
Skin tion methods. Focused efforts to quantify the relative
abundance of Bifidobacterium have determined that this
genus is present as a minority genus in the colon.

Nasal
Human Gut Microbiome, Diet and Probiotics
Human nutritional components and dietary patterns
PC1 (13%) clearly impact microbial composition and presumably
function in the intestine. Children consuming a high-fi-
ber, plant-based diet in western Africa demonstrated a
Fig. 1. The microbiome differs at each body site. This principal
coordinate (PC) plot displays variation in terms of human bacte- relative abundance of two bacterial genera, Prevotella and
rial composition at different body sites. The oral (darkest grey), Xylanibacter, that may contribute to digestion of plant
gastrointestinal (light grey), vaginal (dark grey), nasal (black) and components and fiber in the diet. These two genera con-
skin (grey) microbiomes are shown in different shading. Each tain genes and pathways capable of metabolizing cellulose
point in two-dimensional space represents a different healthy hu- and xylan in the diet, and these genera are rare or absent
man individual and relative distances between the sites indicate
relative differences in bacterial composition (adapted from Hut- in children consuming a westernized diet in southern Eu-
tenhower et al. [12]). rope [17]. A more recent study also described major dif-
ferences in the fecal microbiomes of children in the USA
and Bangladesh [18]. Although fundamental long-term
differences in the diet clearly seem to affect gut microbial
proximate numbers exceeding 50 total human-associated composition, short-term (days) major changes in diet do
bacterial phyla [14, 15]. The composition of the microbi- not have a correspondingly major impact on gastrointes-
ome is distinct at each body site or habitat, each body site tinal microbial composition [19]. It appears that major
has a distinct microbial composition and, presumably, changes in the diet will require longer time periods
differences in function translate to differences in physiol- (months to years) to profoundly shift the composition of
ogy at each body site (e.g. skin and intestine) (fig. 1). the gut microbiome in human individuals. As health care
The relative abundance of probiotic genera and species providers consider probiotic strategies, it is important to
in the healthy human microbiome is a relevant consider- also keep in mind that short-term consumption of probi-
ation, as well as whether microbial deficiencies in indi- otics does not appear to shift microbial composition, but
vidual species could be readily corrected by administra- studies in mouse models suggest that the major impact of
tion of probiotics to children. Alternatively, do probiotics probiotics in the short term may pertain to changes in
simply enhance the ability of other bacterial genera to microbial gene expression and metabolite production
proliferate and reduce the numbers of potentially harm- [20].
ful bacteria? Rational probiotic strategies could be devel- In addition to the effects of diet and probiotic con-
oped that take advantage of predictable changes in micro- sumption on the composition of the gastrointestinal mi-
bial composition following probiotic therapy. For 2 de- crobiome, prebiotics or symbiotic combinations may also
cades, we had scientific evidence that ingestion of affect gut microbial composition. Gibson et al.’s [21]
probiotics in human volunteers resulted in persistence of landmark paper in 1995 provided evidence for the prebi-
probiotic strains (lactobacilli) 11 days later in the small otic concept and the idea that indigestible oligosaccha-
intestine with corresponding reductions in other bacte- rides would provide a substrate for beneficial microbes in
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

44 Ann Nutr Metab 2013;63(suppl 2):42–52 Versalovic


DOI: 10.1159/000354899
Downloaded by:
the intestine. Human milk oligosaccharides (HMOs) sive enzyme indoleamine 2,3-dioxygenase (IDO) [32].
have been characterized in recent years as a distinguish- IDO facilitates the conversion of the amino acid trypto-
ing feature from bovine milk, and these milk components phan to the metabolite kynurenine, and this metabolic
appear to promote the proliferation and biological func- conversion has been associated with virus- and tumor-
tions of probiotic genera such as Bifidobacterium spp. induced immunosuppression. Kynurenine may suppress
[22]. For several decades, it has been recognized that inflammation in the context of a proinflammatory micro-
breast milk-fed infants dem- biome or host condition. Con-
onstrated relative resistance jugated linoleic acids are fatty
to infectious gastroenteritis The human microbiome contains acid derivatives produced by
[23]. The supplementation of an enormous capacity for converting various probiotic species, gen-
probiotics to infant diets nutrients and dietary substrates, erating compounds with anti-
yielded protection against di- inflammatory and anticar-
arrhea and rotaviral infection and this functional capacity cinogenic effects [33]. Probi-
[24]. So, the concepts of pre- can be affected by administration otic Lactobacillus plantarum
biotics, probiotics and effects of probiotic strains. administration in dairy goats
on the resilience and diversity resulted in shifts in gut micro-
of the microbiome become bial composition and altera-
intertwined, and a broader consideration of combina- tions in milk fat composition including greater amounts
tions of probiotics with nutritional strategies may result of polyunsaturated fatty acids such as linoleic acid [34].
in predictable changes to the function of the microbiome. In summary, the human microbiome contains an enor-
The ‘center stage’ importance of nutrition early in life and mous capacity for converting nutrients and dietary sub-
during childhood and adolescent development highlights strates, and this functional capacity can be affected by ad-
the potential impact of probiotics on basic aspects of hu- ministration of probiotic strains.
man nutrition and nutrient availability. Twin pairs in
eastern Africa were differentially susceptible to the condi-
tion of undernutrition known as kwashiorkor, based on Microbiome, Probiotics and Pediatric
differences of the composition of their intestinal micro- Gastroenterology
biomes [25]. Presumably, susceptibilities to the clinical Early Microbial Ecology in the Gut
phenotypes of undernutrition are affected by the meta- The microbial ecology of the human intestine provides
bolic capacity of the microbiome and the abilities of gut an opportunity to understand the nature of microbial
bacteria to convert foodstuffs into available nutrients for populations in the human body and how these popula-
childhood development. tions influence gene expression patterns and the ultimate
Gut bacteria synthesize and convert a variety of com- functionality of the microbiome. It is unclear whether the
pounds that impact the physiology, immunity and pre- fetus is exposed to microbes or their metabolites or DNA,
sumably disease susceptibility or resistance of human in- although investigators are actively exploring the relation-
dividuals. Examples of de novo biosynthesis pathways in ship of the human microbiome to the in utero environ-
the gut microbiome and probiotics include the synthesis ment and mode of delivery [see Luoto et al. in this issue].
of B complex vitamins such as vitamin B12 (cobalamin) During infancy, the composition of the gut microbiome
[26] and vitamin B1 (thiamine) [27]. Examples of luminal fluctuates rapidly [35] and reaches an adult-like equilib-
conversion include the conversion of plant lignins to en- rium by 3 years of age in one study [36] (fig. 2).
terolignins, vitamin K1 (phylloquinone) to vitamin K2
and analogs (menaquinones) [28], amino acid decarbox- Necrotizing Enterocolitis
ylation reactions generating biogenic amines (e.g. histi- The development of the intestinal microbiome in ear-
dine/histamine, glutamate/GABA) [29, 30] and carbohy- ly life and its importance has been highlighted in the pre-
drate/dietary fiber conversion to short chain fatty acids term neonate at risk for developing necrotizing enteroco-
(SCFAs) [31]. With respect to amino acid metabolism, litis (NEC) [see Walker in this issue]. The gut microbi-
oral administration of probiotic Bifidobacterium species ome of infants who developed NEC was characterized by
can stimulate peripheral blood-derived immune cells to compositional differences such as increased abundance
produce greater amounts of the immunosuppressive cy- of γ-Proteobacteria [37], a common feature of the gut mi-
tokine interleukin-10 (IL-10) and the immunosuppres- crobiome in disease states. Compositional differences in
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

The Human Microbiome and Probiotics Ann Nutr Metab 2013;63(suppl 2):42–52 45
DOI: 10.1159/000354899
Downloaded by:
motherapy, so that iatrogenic infections may be corrected
Infants 0–3 years by probiotics. In the past decade, the incidence of pediat-
1,600
ric Clostridium difficile-associated disease has steadily in-
Observed OTUs

1,200
creased [45]. Gorbach et al. [46] demonstrated successful
treatment of C. difficile disease using a single strain of
800 human-derived Lactobacillus rhamnosus (LGG). These
findings laid the foundation for the generalized accep-
400 tance of probiotics in the last decade of the twentieth cen-
tury. This strain of interest became commonly known as
0 0.3 0.7 1.1 1.5 1.9 2.3 2.7 3.0 the probiotic LGG and has been applied in numerous pe-
Age (years) diatric studies [47, 48]. Several studies in children have
demonstrated that probiotics may be effective at sup-
pressing antibiotic-associated diarrhea [49, 50], and pro-
Fig. 2. Bacterial diversity increases with age. Intestinal bacterial
composition increases steadily during the first 3 years of life. Stan-
biotics may promote restoration of microbial diversity as
dard errors of the mean are plotted (adapted with permission from one mechanism for amelioration of the disease pheno-
MacMillan Publishers Ltd. from Yatsunenko et al. [36]). Black cir- type [51]. Prior evidence showed that the human intesti-
cles = US; grey circles = Amerindians; white circles = Malawians; nal microbiome was restricted in terms of bacterial diver-
OTU = operational taxonomic unit (bacterial taxon). sity in patients with recurrent C. difficile disease [52]. Pre-
sumably, a gut microbiome with limited diversity (an
example of dysbiosis) creates a permissive environment
the fecal microbiota preceded the development of NEC. of recurrent colitis due to C. difficile, and probiotics may
Possibly, probiotics or nutritional approaches could shift be useful by promoting increased gut bacterial diversity.
microbial composition towards a more disease-resistant The success of fecal microbiota or intestinal microbiome
gut microbiome. A diet composed entirely of human milk transplantation in C. difficile-associated disease [53] pro-
has been effective in reducing the incidence of NEC in vides additional evidence that restoration of sufficient
premature infants, and donor human milk supplementa- bacterial diversity and functional capacity can effectively
tion has been recommended as one strategy to prevent treat this disorder of microbial ecology.
NEC [38, 39]. In addition to human milk supplementa- The American Academy of Pediatrics (AAP) endorsed
tion, several probiotics have yielded success in the pre- the application of probiotics for the prevention of antibi-
vention of severe NEC and all-cause mortality in prema- otic-associated diarrhea and the treatment of acute viral
ture infants, including very-low-birth-weight infants [40, gastroenteritis in healthy children [54]. Although data are
41]. Data could not be extrapolated to extremely-low- lacking with respect to changes in the intestinal microbi-
birth-weight infants. Heterogeneity among the clinical ome in cases of acute bacterial or viral gastroenteritis in
trials in terms of design and probiotic strains prevents any humans, probiotics have demonstrated their ability to
firm recommendation regarding a specific probiotic shorten the course of disease and ameliorate symptoms
strain for prevention of NEC [42]. An increased incidence in several studies spanning 2 decades [49, 55]. The addi-
of NEC has been associated with administration of hista- tion of probiotics may stimulate the mucosal immune
mine 2 receptor (H2R) antagonists as acid blockers to system and the microbiome’s own defense mechanisms,
preterm infants [43, 44]. The histamine signaling path- resulting in rapid pathogen clearance and mucosal heal-
way may provide an opportunity for targeted interven- ing.
tions of probiotics based on known mechanisms. Probi-
otics capable of converting the dietary amino acid L-his- Celiac Disease
tidine to histamine have been reported [29], and histamine Although the microbial composition of the small in-
may suppress inflammation by promoting H2R signaling testine does not appear to differ in patients with celiac
in the intestinal mucosa. disease, differences in the intestinal microbiome were de-
tected in self-collected stool specimens obtained from pa-
Recurrent Clostridium difficile Infection and Acute tients with celiac disease [56]. Corresponding changes in
Gastroenteritis the fecal microbiome and the fecal and urinary metabo-
Disorders of microbial ecology may be caused by con- lome were reported in children with celiac disease com-
sumption of antimicrobial agents (antibiotics) and che- pared to healthy controls [56]. Breast milk feeding with
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

46 Ann Nutr Metab 2013;63(suppl 2):42–52 Versalovic


DOI: 10.1159/000354899
Downloaded by:
its possible ‘feeder’ effects on beneficial microbes and dobacteria may explain the relative success of a Bifidobac-
HLA genotype were found to influence the relative sus- terium-Lactobacillus combination strategy [68] versus a
ceptibilities to celiac disease in the PROFICEL study [57]. Lactobacillus-only probiotics strategy [69] in children
In a provocative report, the relative timing of gluten in- with IBS.
troduction in early childhood appeared to impact disease A general consensus in the field is that many function-
susceptibility, and the disease phenotypes were correlated al gastrointestinal disorders including IBS can best be un-
with changes in the intestinal microbiome and metabo- derstood as disorders of brain-gut interactions [70]. The
lome [58]. An interesting aspect relevant to celiac disease brain-gut axis represents a bidirectional connection be-
is the presence of gluten-metabolizing bacterial genera tween the digestive and nervous systems with emerging
such as Rothia spp. in the oral microbiome [59]. Gluten- importance to human biology and medicine [70]. Recent
metabolizing microbes in the preclinical evidence suggests
oral or intestinal microbiomes that changes in the composi-
may reduce the relative sus- Changes in the composition and tion and function of the mam-
ceptibilities of individuals function of the mammalian gut malian gut microbiome can
with a genetic predisposition
microbiome can affect brain systems affect brain systems related to
to celiac disease due to HLA pain and affect regulation
gene polymorphisms. Future related to pain and affect regulation. [71]. In rodents, the lack of
probiotic strategies may in- gut microbes in germ-free
clude consideration of probi- mice [72, 73] and the modula-
otics with gluten metabolism genes and the ability of pro- tion of gut microbial ecology by probiotics [74] and anti-
biotics to enhance the function of such gluten-metaboliz- biotics [75] have been associated with changes in affective
ing bacteria in the microbiome. behavior, pain responses and gene expression in the
brain. The production of a variety of neuroactive signal-
Irritable Bowel Syndrome ing molecules by bacterial components of the microbi-
Differences in composition or dysbiosis of the gut mi- ome provides additional evidence that the gut microbi-
crobiome in irritable bowel syndrome (IBS) were first re- ome may generate signals with remote effects in different
ported with comprehensive DNA sequencing and array organ systems, including the central nervous system. Re-
studies in 2011 [60, 61]. Disease signatures based on dif- cent preclinical data in rodents suggest that changes in
ferences in bacterial composition were detected and dis- the gut microbiota can be associated with changes in the
tinguished patients with more frequent abdominal pain expression of brain signaling systems and associated
and more severe gastrointestinal disease phenotypes. emotional behavior [72, 74], and oral administration of
Overlapping features included the enrichment of probiotics to healthy women demonstrated changes in in-
γ-Proteobacteria in children and adults with IBS, and an teroceptive, affective and reward circuits in response to
association of this group containing known enteric patho- chronic probiotic ingestion [76]. In summary, probiotics
gens with increased pain symptoms. Subsequent studies may be useful for the prevention or treatment of func-
confirmed and extended these findings regarding distinct tional gastrointestinal disorders like IBS by affecting the
compositional differences of the intestinal microbiome in function of the gut microbiome or by altering brain func-
IBS [62–64]. In children, a follow-up study described dif- tion and pain perception centrally.
ferences in the fecal microbiome such as relative deficien-
cies of the genera Bifidobacterium and Verrucomicrobium Inflammatory Bowel Disease
in children with IBS-diarrheal predominant subtype Differences in the composition of the intestinal micro-
(IBS-D) [65]. Administration of Bifidobacterium spp. in biome have been reported in several studies of patients
adult patients with IBS reduced symptoms and highlight- with Crohn’s disease and ulcerative colitis. Such differ-
ed the potential benefits of probiotic therapies in IBS with ences include reduced proportions of the bacterial phyla
carefully selected probiotic strains [66, 67]. Reduced Bacteroidetes and Firmicutes, relative deficiencies of the
amounts of Bifidobacterium spp. in the intestinal micro- genus Faecalibacterium in ileal Crohn’s disease and ex-
biomes of children and adults with IBS point towards a pansion of the phylum Proteobacteria in patients with in-
rational basis for supplementation of ‘missing’ or defi- flammatory bowel disease (IBD) [77–79]. Early successes
cient bacteria in disease conditions as a way to prevent or with probiotics in the context of acute and chronic pou-
treat disease. The suggested importance of intestinal Bifi- chitis [80, 81] have been followed with mixed results and
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

The Human Microbiome and Probiotics Ann Nutr Metab 2013;63(suppl 2):42–52 47
DOI: 10.1159/000354899
Downloaded by:
disappointments in clinical trials of probiotics for the ture. Unfortunately for patients, the identification of pro-
treatment of IBD [82]. The relative enrichment of Proteo- biotic strains that bestow beneficial effects on the human
bacteria and specifically γ-Proteobacteria in recent stud- skin has not been defined, and such applications in der-
ies emphasizes the potential importance of Gram-nega- matology await further investigation. Past limited suc-
tive bacteria in adult and pediatric IBD. Specific compo- cesses with oral probiotics and amelioration of atopic skin
nents including γ-Proteobacteria were useful for identifi- disease features in children [88, 89] have generated opti-
cation of children with IBD; a specific example was the mism for the potential roles of oral or topical probiotics
enrichment of the genus Escherichia/Shigella in children in the treatment of atopy. However, this enthusiasm has
with ulcerative colitis [83]. These findings are relevant been tempered by the realization that many gaps exist in
because the genus Escherichia coli has been the source of our knowledge of the skin microbiome, probiotics and
an established probiotic strain in humans, and microbi- pediatric allergic diseases, and no single probiotic strain
ome research may help steer physicians towards optimal can be recommended at this time [90].
probiotic/disease combinations. Recent advances in
terms of understanding functional metagenomics may Relative differences in composition
point to the next generation of probiotics for adult and
pediatric IBD. Microbial function was more often affect-
and function of bacterial communities
ed than microbial composition in a population of adult on the human skin may explain
patients with Crohn’s disease and ulcerative colitis [79]. different patterns of atopic diseases
Major shifts in oxidative stress were identified in the adult involving the skin and airways.
IBD state, and relative reductions were identified in genes
and pathways involved in carbohydrate metabolism and
amino acid biosynthesis in IBD [79]. These differences in
terms of metagenomic capacity may be important for the Microbiome of the Airways: Asthma and Atopy
rational selection of probiotics supplying ‘missing’ func- Alterations in the human microbiome and pathogens
tions or factors that interfere with disease-promoting have been implicated as possible causes of asthma and
pathways in the microbiome. potential triggers of asthmatic episodes. In a study of
healthy children and children with asthma, there were no
significant shifts in bacterial phyla detected in the respira-
Microbiome, Probiotics and Atopic Disease tory tract, and the predominant phyla in both groups in-
Microbiome of the Human Skin and Atopy cluded Bacteroidetes, Firmicutes and Proteobacteria [91].
The human skin contains several dominant bacterial Whereas healthy children were characterized by the gen-
genera across different sites, including Corynebacterium, era Prevotella, Streptococcus, Veillonella and Fusobacte-
Eubacterium, Propionibacterium, Staphylococcus and rium, the genus Haemophilus was relatively abundant in
Streptococcus [84], and one dominant fungal genus Mal- the asthmatic group. Within the genus Haemophilus, the
assezia [85]. Focused studies on specific body compart- pathogenic species Haemophilus influenzae was previ-
ments have highlighted key features of colonization in ously implicated as a potential trigger of asthmatic epi-
healthy individuals. Corynebacterium was the most com- sodes. A pediatric study from Ecuador yielded intriguing
mon bacterial genus in the anterior nares [86], and the results with respect to the airways microbiome; treatment
human pathogen Staphylococcus aureus was present in a of respiratory illnesses differs greatly in Ecuador from the
substantial proportion (36%) of healthy human subjects standard of care in the United States. Oropharyngeal
[87]. Relative differences in composition and function of swabs were obtained from wheezing and healthy infants,
bacterial communities on the human skin may explain and all patients had minimal exposure to antibiotics and
different patterns of atopic diseases involving the skin no exposure to inhaled steroids [92]. The overall bacte-
and airways. In cases of atopic dermatitis, staphylococci rial community in the study population (healthy and
including S. aureus and S. epidermidis populations appear wheezing children) consisted primarily of the bacterial
to ‘bloom’ and contribute to disease flares and relapse at phyla Firmicutes, Proteobacteria, Actinobacteria, Bacte-
specific skin sites [87]. Perhaps, the topical application of roidetes and Fusobacterium in order of predominance.
probiotics or skin bacterial communities that suppress The most common genera isolated were consistent with
pathogen ‘blooms’ on specific body surfaces may help a prior study [91], with most bacteria belonging to Strep-
prevent or mitigate these atopic disease flares in the fu- tococcus, Veillonella, Atopobium and Prevotella. In the
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

48 Ann Nutr Metab 2013;63(suppl 2):42–52 Versalovic


DOI: 10.1159/000354899
Downloaded by:
Fig. 3. Probiotics modulate key signaling
pathways in intestinal epithelial cells. Dif-
ferent probiotic species and strains target
distinct signaling pathways in the gut mu-
cosa. These pathways may be exploited to
select specific probiotics based on defined
mechanisms of action affecting the intesti-
nal microbiome and the mucosal immune
system (adapted with permission from
MacMillan Publishers Ltd. on behalf of
Cander Research UK from Thomas and
Versalovic [93]).

wheezing group, a greater frequency of the bacterial gen- troenteritis and IBS. An enhanced understanding of the
era Neisseria, Corynebacterium, Staphylococcus, Actino- effects of probiotics on the microbiome should facilitate
myces and Haemophilus were identified [92]. Possibly, selection of optimal probiotic strains for specific diseases.
these differences in the microbiome of the airways ac- Future directions include studies of effects of specific
count for differences in the susceptibility to asthma or probiotic strains on the human microbiome. Such studies
symptoms such as wheezing in the context of immune may include experiments evaluating changes in micro-
dysregulation. To reiterate, although future probiotic bial composition using in vitro model systems, ‘human-
strategies may be applied by oral or inhaled administra- ized’ animal models containing human-associated bacte-
tion, many gaps exist in our knowledge about the micro- ria, and clinical studies determining effects on human-
biome of the airways, effective probiotics and effects on associated bacterial communities following probiotics
asthma and allergic diseases [90]. administration. Changes in composition could be ex-
tended to evaluation of changes in microbiome function
and the related changes in specific metabolic pathways
Summary and Future Directions caused by individual probiotic strains. By understanding
Steady advances in our knowledge of the composition how probiotic strains alter specific functions of the hu-
and function of the human microbiome at multiple body man microbiome at different body sites, probiotic strain
sites including the gut, skin and airways should contribute selection may be optimized for specific disease states
to our understanding of mechanisms of probiosis. Al- (fig. 3). As we proceed into the era of metagenomic med-
though our knowledge about microbial composition in icine, patients may be tested for their own microbial com-
Homo sapiens is progressing rapidly, many gaps exist in positional and functional features so that probiotics may
our knowledge about the functional capacity and meta- be customized and tailored to the disease state and the
bolic machinery of the human microbiome. Although individual patient. The fusion of the microbiome with
more studies are needed, probiotics appear capable of af- microbe-based therapies in medicine will advance the
fecting the composition and function of the microbiome. causes of holistic and personalized medicine.
Effects on function are likely to be more important in the
short term (hours to days) following initial administra-
tion. Probiotics have yielded beneficial effects in numer- Disclosure Statement
ous studies in the context of different disease states in Dr. Versalovic receives unrestricted research support from
pediatric gastroenterology. These disease states include BioGaia AB. The writing of this article was supported by Nestlé
NEC, antibiotic-associated diarrhea and colitis, acute gas- Nutrition Institute.
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

The Human Microbiome and Probiotics Ann Nutr Metab 2013;63(suppl 2):42–52 49
DOI: 10.1159/000354899
Downloaded by:
References
1 Pirisi A: Phage therapy – advantages over an- 14 Rappe MS, Giovannoni SJ: The uncultured 26 Santos F, Vera JL, van der Heijden R, Valdez
tibiotics? Lancet 2000;356:1418. microbial majority. Annu Rev Microbiol G, de Vos WM, Sesma F, Hugenholtz J: The
2 Keen EC: Phage therapy: concept to cure. 2003;57:369–394. complete coenzyme B12 biosynthesis gene
Front Microbiol 2012; 3:238. 15 Pace NR: Mapping the tree of life: progress cluster of Lactobacillus reuteri CRL1098. Mi-
3 Fleming A: On the antibacterial action of and prospects. Microbiol Mol Biol Rev 2009; crobiology 2008;154:81–93.
cultures of a penicillium, with special refer- 73:565–576. 27 Saulnier DM, Santos F, Roos S, Mistretta TA,
ence to their use in the isolation of B. influ- 16 Johansson ML, Molin G, Jeppsson B, Nobaek Spinler JK, Molenaar D, Teusink B, Versa-
enzae (reprinted from Br J Exp Pathol). Clin S, Ahrne S, Bengmark S: Administration of lovic J: Exploring metabolic pathway recon-
Infect Dis 1980;2:129–139. different Lactobacillus strains in fermented struction and genome-wide expression pro-
4 Metchnikoff I: The Prolongation of Life: Op- oatmeal soup: in vivo colonization of human filing in Lactobacillus reuteri to define func-
timistic Studies. New York, Springer Pub- intestinal mucosa and effect on the indigenous tional probiotic features. PLoS One 2011;
lishing Company, 1908. flora. Appl Environ Microbiol 1993;59:15–20. 6:e18783.
5 Lilly DM, Stillwell RH: Probiotics: growth- 17 De Filippo C, Cavalieri D, Di Paola M, 28 Dairi T: Menaquinone biosyntheses in mi-
promoting factors produced by microorgan- Ramazzotti M, Poullet JB, Massart S, Collini croorganisms. Methods Enzymol 2012; 515:
isms. Science 1965;147:747–748. S, Pieraccini G, Lionetti P: Impact of diet in 107–122.
6 Parker RB: Probiotics, the other half of the shaping gut microbiota revealed by a com- 29 Thomas CM, Hong T, van Pijkeren JP, He-
antibiotic story. Anim Nutr Health 1974;29: parative study in children from Europe and marajata P, Trinh DV, Hu W, Britton RA,
4–8. rural Africa. Proc Natl Acad Sci USA 2010; Kalkum M, Versalovic J: Histamine derived
7 Fuller R: Probiotics in man and animals. J 107:14691–14696. from probiotic Lactobacillus reuteri sup-
Appl Bacteriol 1989;66:365–378. 18 Lin A, Bik EM, Costello EK, Dethlefsen L, presses TNF via modulation of PKA and
8 FAO/WHO: Health and Nutritional Proper- Haque R, Relman DA, Singh U: Distinct dis- ERK signaling. PLoS One 2012;7:e31951.
ties of Probiotics in Food Including Powder tal gut microbiome diversity and composi- 30 De Biase D, Pennacchietti E: Glutamate decar-
Milk with Live Lactic Acid Bacteria. Cordo- tion in healthy children from Bangladesh boxylase-dependent acid resistance in orally
ba, 1–4 October, 2001. and the United States. PLoS One 2013; acquired bacteria: function, distribution and
9 Peterson J, Garges S, Giovanni M, McInnes P, 8:e53838. biomedical implications of the gadBC operon.
Wang L, Schloss JA, Bonazzi V, McEwen JE, 19 Wu GD, Chen J, Hoffmann C, Bittinger K, Mol Microbiol 2012;86:770–786.
Wetterstrand KA, Deal C, Baker CC, Di Fran- Chen YY, Keilbaugh SA, Bewtra M, Knights 31 van Zanten GC, Knudsen A, Roytio H,
cesco V, Howcroft TK, Karp RW, Lunsford D, Walters WA, Knight R, Sinha R, Gilroy E, Forssten S, Lawther M, Blennow A, Lahtinen
RD, Wellington CR, Belachew T, Wright M, Gupta K, Baldassano R, Nessel L, Li H, Bush- SJ, Jakobsen M, Svensson B, Jespersen L: The
Giblin C, David H, Mills M, Salomon R, Mul- man FD, Lewis JD: Linking long-term di- effect of selected synbiotics on microbial
lins C, Akolkar B, Begg L, Davis C, Grandison etary patterns with gut microbial entero- composition and short-chain fatty acid pro-
L, Humble M, Khalsa J, Little AR, Peavy H, types. Science 2011;334:105–108. duction in a model system of the human co-
Pontzer C, Portnoy M, Sayre MH, Starke- 20 McNulty NP, Yatsunenko T, Hsiao A, Faith lon. PLoS One 2012;7:e47212.
Reed P, Zakhari S, Read J, Watson B, Guyer JJ, Muegge BD, Goodman AL, Henrissat B, 32 Konieczna P, Groeger D, Ziegler M, Frei R,
M: The NIH human microbiome project. Ge- Oozeer R, Cools-Portier S, Gobert G, Cher- Ferstl R, Shanahan F, Quigley EM, Kiely B,
nome Res 2009;19:2317–2323. vaux C, Knights D, Lozupone CA, Knight R, Akdis CA, O’Mahony L: Bifidobacterium in-
10 Johnson CL, Versalovic J: The human micro- Duncan AE, Bain JR, Muehlbauer MJ, New- fantis 35624 administration induces Foxp3 T
biome and its potential importance to pedi- gard CB, Heath AC, Gordon JI: The impact regulatory cells in human peripheral blood:
atrics. Pediatrics 2012;129:950–960. of a consortium of fermented milk strains on potential role for myeloid and plasmacytoid
11 Methe BA, Nelson KE, Pop M, Creasy HH, the gut microbiome of gnotobiotic mice and dendritic cells. Gut 2012;61:354–366.
Giglio MG, Huttenhower C, Gevers D, Petro- monozygotic twins. Sci Transl Med 2011; 3: 33 Ewaschuk JB, Walker JW, Diaz H, Madsen
sino J, Abubucker S, Badger JH, Chinwalla 106ra. KL: Bioproduction of conjugated linoleic
AT, Ear AM, FitzGerald MG, Fulton RS, 21 Gibson GR, Beatty ER, Wang X, Cummings acid by probiotic bacteria occurs in vitro and
Hallsworth-Pepin K, Lobos EA, Madupu R, JH: Selective stimulation of bifidobacteria in in vivo in mice. J Nutr 2006;136:1483–1487.
Magrini V, Martin JC, Mitreva M, Muzny the human colon by oligofructose and inu- 34 Maragkoudakis PA, Mountzouris KC, Rosu
DM, Sodergren EJ, Versalovic J, et al: A lin. Gastroenterology 1995; 108:975–982. C, Zoumpopoulou G, Papadimitriou K, Dal-
framework for human microbiome research. 22 Bode L: Human milk oligosaccharides: every aka E, Hadjipetrou A, Theofanous G, Strozzi
Nature 2012;486:215–221. baby needs a sugar mama. Glycobiology GP, Carlini N, Zervas G, Tsakalidou E: Feed
12 Huttenhower C, Gevers D, Knight R, Abu- 2012;22:1147–1162. supplementation of Lactobacillus plantarum
bucker S, Badger JH, Chinwalla AT, Creasy 23 Bullen CL, Willis AT: Resistance of the PCA 236 modulates gut microbiota and milk
HH, Earl AM, FitzGerald MG, Fulton RS, breast-fed infant to gastroenteritis. Br Med J fatty acid composition in dairy goats – a pre-
Giglio MG, Hallsworth-Pepin K, Lobos EA, 1971;3:338–343. liminary study. Int J Food Microbiol 2010;
Madupu R, Magrini V, Martin JC, Mitreva 24 Saavedra JM, Bauman NA, Oung I, Perman 141(suppl 1):S109–S116.
M, Muzny D, Sodergren E, Versalovic J, Wol- JA, Yolken RH: Feeding of Bifidobacterium 35 Palmer C, Bik EM, DiGiulio DB, Relman DA,
lam AM, Worley KC, Wortman JR, Young bifidum and Streptococcus thermophilus to Brown PO: Development of the human in-
SK, Zeng Q, Aagaard K, et al: Structure, infants in hospital for prevention of diar- fant intestinal microbiota. PLoS Biol 2007;
function and diversity of the healthy human rhoea and shedding of rotavirus. Lancet 5:e177.
microbiome. Nature 2012;486:207–214. 1994;344:1046–1049. 36 Yatsunenko T, Rey FE, Manary MJ, Trehan I,
13 Zhou Y, Gao H, Mihindukulasuriya KA, La 25 Smith MI, Yatsunenko T, Manary MJ, Trehan Dominguez-Bello MG, Contreras M, Magris
Rosa PS, Wylie KM, Vishnivetskaya T, Podar I, Mkakosya R, Cheng J, Kau AL, Rich SS, M, Hidalgo G, Baldassano RN, Anokhin AP,
M, Warner B, Tarr PI, Nelson DE, Forten- Concannon P, Mychaleckyj JC, Liu J, Houpt Heath AC, Warner B, Reeder J, Kuczynski J,
berry JD, Holland MJ, Burr SE, Shannon E, Li JV, Holmes E, Nicholson J, Knights D, Caporaso JG, Lozupone CA, Lauber C, Cle-
WD, Sodergren E, Weinstock GM: Biogeog- Ursell LK, Knight R, Gordon JI: Gut microbi- mente JC, Knights D, Knight R, Gordon JI:
raphy of the ecosystems of the healthy hu- omes of Malawian twin pairs discordant for Human gut microbiome viewed across age
man body. Genome Biol 2013; 14:R1. kwashiorkor. Science 2013; 339:548–554. and geography. Nature 2012;486:222–227.
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

50 Ann Nutr Metab 2013;63(suppl 2):42–52 Versalovic


DOI: 10.1159/000354899
Downloaded by:
37 Mai V, Young CM, Ukhanova M, Wang X, 48 Guandalini S: Probiotics for prevention and 59 Fernandez-Feo M, Wei G, Blumenkranz G,
Sun Y, Casella G, Theriaque D, Li N, Sharma treatment of diarrhea. J Clin Gastroenterol Dewhirst FE, Schuppan D, Oppenheim FG,
R, Hudak M, Neu J: Fecal microbiota in pre- 2011;45(suppl):S149–S153. Helmerhorst EJ: The cultivable human oral
mature infants prior to necrotizing entero- 49 Preidis GA, Versalovic J: Targeting the hu- gluten-degrading microbiome and its poten-
colitis. PLoS One 2011;6:e20647. man microbiome with antibiotics, probiot- tial implications in coeliac disease and glu-
38 Sullivan S, Schanler RJ, Kim JH, Patel AL, ics, and prebiotics: gastroenterology enters ten sensitivity. Clin Microbiol Infect 2013;
Trawoger R, Kiechl-Kohlendorfer U, Chan the metagenomics era. Gastroenterology 19:E386–E394.
GM, Blanco CL, Abrams S, Cotten CM, La- 2009;136:2015–2031. 60 Saulnier DM, Riehle K, Mistretta TA, Diaz
roia N, Ehrenkranz RA, Dudell G, Cristofalo 50 Goldenberg JZ, Ma SS, Saxton JD, Martzen MA, Mandal D, Raza S, Weidler EM, Qin X,
EA, Meier P, Lee ML, Rechtman DJ, Lucas A: MR, Vandvik PO, Thorlund K, Guyatt GH, Coarfa C, Milosavljevic A, Petrosino JF,
An exclusively human milk-based diet is as- Johnston BC: Probiotics for the prevention of Highlander S, Gibbs R, Lynch SV, Shulman
sociated with a lower rate of necrotizing en- Clostridium difficile-associated diarrhea in RJ, Versalovic J: Gastrointestinal microbi-
terocolitis than a diet of human milk and bo- adults and children. Cochrane Database Syst ome signatures of pediatric patients with ir-
vine milk-based products. J Pediatr 2010; Rev 2013;5:CD006095. ritable bowel syndrome. Gastroenterology
156:562–567.e1. 51 Preidis GA, Saulnier DM, Blutt SE, Mistretta 2011;141:1782–1791.
39 Fallon EM, Nehra D, Potemkin AK, Gura TA, Riehle KP, Major AM, Venable SF, Bar- 61 Rajilic-Stojanovic M, Biagi E, Heilig HG, Ka-
KM, Simpser E, Compher C, Puder M: AS- rish JP, Finegold MJ, Petrosino JF, Guerrant jander K, Kekkonen RA, Tims S, de Vos WM:
PEN clinical guidelines: nutrition support of RL, Conner ME, Versalovic J: Host response Global and deep molecular analysis of mi-
neonatal patients at risk for necrotizing en- to probiotics determined by nutritional sta- crobiota signatures in fecal samples from pa-
terocolitis. JPEN J Parenter Enteral Nutr tus of rotavirus-infected neonatal mice. J Pe- tients with irritable bowel syndrome. Gas-
2012;36:506–523. diatr Gastroenterol Nutr 2012; 55:299–307. troenterology 2011;141:1792–1801.
40 Alfaleh K, Anabrees J, Bassler D, Al-Kharfi 52 Chang JY, Antonopoulos DA, Kalra A, 62 Jeffery IB, O’Toole PW, Ohman L, Claesson
T: Probiotics for prevention of necrotizing Tonelli A, Khalife WT, Schmidt TM, Young MJ, Deane J, Quigley EM, Simren M: An ir-
enterocolitis in preterm infants. Cochrane VB: Decreased diversity of the fecal microbi- ritable bowel syndrome subtype defined by
Database Syst Rev 2011;3:CD005496. ome in recurrent Clostridium difficile-asso- species-specific alterations in faecal micro-
41 Mihatsch WA, Braegger CP, Decsi T, Kolacek ciated diarrhea. J Infect Dis 2008; 197: 435– biota. Gut 2012;61:997–1006.
S, Lanzinger H, Mayer B, Moreno LA, Poh- 438. 63 Carroll IM, Ringel-Kulka T, Siddle JP, Ringel
landt F, Puntis J, Shamir R, Stadtmuller U, 53 Mattila E, Uusitalo-Seppala R, Wuorela M, Y: Alterations in composition and diversity
Szajewska H, Turck D, van Goudoever JB: Lehtola L, Nurmi H, Ristikankare M, of the intestinal microbiota in patients with
Critical systematic review of the level of evi- Moilanen V, Salminen K, Seppala M, Mattila diarrhea-predominant irritable bowel syn-
dence for routine use of probiotics for reduc- PS, Anttila VJ, Arkkila P: Fecal transplanta- drome. Neurogastroenterol Motil 2012; 24:
tion of mortality and prevention of necrotiz- tion, through colonoscopy, is effective ther- 521–530, e248.
ing enterocolitis and sepsis in preterm in- apy for recurrent Clostridium difficile infec- 64 Chassard C, Dapoigny M, Scott KP, Crouzet
fants. Clin Nutr 2012;31:6–15. tion. Gastroenterology 2012; 142:490–496. L, Del’homme C, Marquet P, Martin JC,
42 Downard CD, Renaud E, St Peter SD, Abdul- 54 Thomas DW, Greer FR: Probiotics and pre- Pickering G, Ardid D, Eschalier A, Dubray
lah F, Islam S, Saito JM, Blakely ML, Huang biotics in pediatrics. Pediatrics 2010; 126: C, Flint HJ, Bernalier-Donadille A: Func-
EY, Arca MJ, Cassidy L, Aspelund G: Treat- 1217–1231. tional dysbiosis within the gut microbiota of
ment of necrotizing enterocolitis: an Ameri- 55 Piescik-Lech M, Shamir R, Guarino A, Sza- patients with constipated-irritable bowel
can Pediatric Surgical Association outcomes jewska H: Review article: the management of syndrome. Aliment Pharmacol Ther 2012;
and clinical trials committee systematic re- acute gastroenteritis in children. Aliment 35:828–838.
view. J Pediatr Surg 2012;47:2111–2122. Pharmacol Ther 2013;37:289–303. 65 Rigsbee L, Agans R, Shankar V, Kenche H,
43 Guillet R, Stoll BJ, Cotten CM, Gantz M, Mc- 56 Di Cagno R, De Angelis M, De Pasquale I, Khamis HJ, Michail S, Paliy O: Quantitative
Donald S, Poole WK, Phelps DL: Association Ndagijimana M, Vernocchi P, Ricciuti P, Ga- profiling of gut microbiota of children with
of H2-blocker therapy and higher incidence gliardi F, Laghi L, Crecchio C, Guerzoni ME, diarrhea-predominant irritable bowel syn-
of necrotizing enterocolitis in very low birth Gobbetti M, Francavilla R: Duodenal and drome. Am J Gastroenterol 2012; 107: 1740–
weight infants. Pediatrics 2006; 117:e137– faecal microbiota of celiac children: molecu- 1751.
e142. lar, phenotype and metabolome character- 66 O’Mahony L, McCarthy J, Kelly P, Hurley G,
44 Terrin G, Passariello A, De Curtis M, Man- ization. BMC Microbiol 2011;11:219. Luo F, Chen K, O’Sullivan GC, Kiely B, Col-
guso F, Salvia G, Lega L, Messina F, Paludet- 57 Palma GD, Capilla A, Nova E, Castillejo G, lins JK, Shanahan F, Quigley EM: Lactoba-
to R, Canani RB: Ranitidine is associated Varea V, Pozo T, Garrote JA, Polanco I, Lopez cillus and bifidobacterium in irritable bowel
with infections, necrotizing enterocolitis, A, Ribes-Koninckx C, Marcos A, Garcia-No- syndrome: symptom responses and relation-
and fatal outcome in newborns. Pediatrics vo MD, Calvo C, Ortigosa L, Pena-Quintana ship to cytokine profiles. Gastroenterology
2012;129:e40–e45. L, Palau F, Sanz Y: Influence of milk-feeding 2005;128:541–551.
45 Kim J, Smathers SA, Prasad P, Leckerman type and genetic risk of developing coeliac 67 Whorwell PJ, Altringer L, Morel J, Bond Y,
KH, Coffin S, Zaoutis T: Epidemiological disease on intestinal microbiota of infants: Charbonneau D, O’Mahony L, Kiely B, Sha-
features of Clostridium difficile-associated the PROFICEL study. PLoS One 2012; nahan F, Quigley EM: Efficacy of an encapsu-
disease among inpatients at children’s hospi- 7:e30791. lated probiotic Bifidobacterium infantis 35624
tals in the United States, 2001–2006. Pediat- 58 Sellitto M, Bai G, Serena G, Fricke WF, Stur- in women with irritable bowel syndrome. Am
rics 2008;122:1266–1270. geon C, Gajer P, White JR, Koenig SS, Saka- J Gastroenterol 2006;101:1581–1590.
46 Gorbach SL, Chang TW, Goldin B: Success- moto J, Boothe D, Gicquelais R, Kryszak D, 68 Guandalini S, Magazzu G, Chiaro A, La Bal-
ful treatment of relapsing Clostridium diffi- Puppa E, Catassi C, Ravel J, Fasano A: Proof estra V, Di Nardo G, Gopalan S, Sibal A, Ro-
cile colitis with Lactobacillus GG. Lancet of concept of microbiome-metabolome anal- mano C, Canani RB, Lionetti P, Setty M:
1987;2:1519. ysis and delayed gluten exposure on celiac VSL#3 improves symptoms in children with
47 Gorbach SL: Probiotics and gastrointestinal disease autoimmunity in genetically at-risk irritable bowel syndrome: a multicenter, ran-
health. Am J Gastroenterol 2000; 95(suppl infants. PLoS One 2012;7:e33387. domized, placebo-controlled, double-blind,
1):S2–S4. crossover study. J Pediatr Gastroenterol Nutr
2010;51:24–30.
Universidade Federal do Rio de Janeiro
146.164.3.22 - 1/27/2014 4:15:39 AM

The Human Microbiome and Probiotics Ann Nutr Metab 2013;63(suppl 2):42–52 51
DOI: 10.1159/000354899
Downloaded by:
69 Bausserman M, Michail S: The use of Lacto- 78 Sokol H, Seksik P, Furet JP, Firmesse O, Montemayor C, Mullikin J, Riebow N,
bacillus GG in irritable bowel syndrome in Nion-Larmurier I, Beaugerie L, Cosnes J, Schandler K, Schmidt B, Sison C, Stantripop
children: a double-blind randomized control Corthier G, Marteau P, Dore J: Low counts of M, Thomas J, Thomas P, Vemulapalli M,
trial. J Pediatr 2005;147:197–201. Faecalibacterium prausnitzii in colitis mi- Young A, Kong HH, Segre JA: Topographic
70 Mayer EA: Gut feelings: the emerging biolo- crobiota. Inflamm Bowel Dis 2009;15:1183– diversity of fungal and bacterial communi-
gy of gut-brain communication. Nat Rev 1189. ties in human skin. Nature 2013, E-pub
Neurosci 2011;12:453–466. 79 Morgan XC, Tickle TL, Sokol H, Gevers D, ahead of print.
71 Cryan JF, Dinan TG: Mind-altering micro- Devaney KL, Ward DV, Reyes JA, Shah SA, 86 Grice EA, Segre JA: The skin microbiome.
organisms: the impact of the gut microbiota LeLeiko N, Snapper SB, Bousvaros A, Korze- Nat Rev Microbiol 2011;9:244–253.
on brain and behaviour. Nat Rev Neurosci nik J, Sands BE, Xavier RJ, Huttenhower C: 87 Kong HH, Oh J, Deming C, Conlan S, Grice
2012;13:701–712. Dysfunction of the intestinal microbiome in EA, Beatson MA, Nomicos E, Polley EC, Ko-
72 Diaz Heijtz R, Wang S, Anuar F, Qian Y, inflammatory bowel disease and treatment. marow HD, Murray PR, Turner ML, Segre
Bjorkholm B, Samuelsson A, Hibberd ML, Genome Biol 2012; 13:R79. JA: Temporal shifts in the skin microbiome
Forssberg H, Pettersson S: Normal gut mi- 80 Gionchetti P, Rizzello F, Venturi A, Brigidi P, associated with disease flares and treatment
crobiota modulates brain development and Matteuzzi D, Bazzocchi G, Poggioli G, Mi- in children with atopic dermatitis. Genome
behavior. Proc Natl Acad Sci USA 2011; 108: glioli M, Campieri M: Oral bacteriotherapy Res 2012;22:850–859.
3047–3052. as maintenance treatment in patients with 88 Majamaa H, Isolauri E: Probiotics: a novel
73 Amaral FA, Sachs D, Costa VV, Fagundes chronic pouchitis: a double-blind, placebo- approach in the management of food aller-
CT, Cisalpino D, Cunha TM, Ferreira SH, controlled trial. Gastroenterology 2000; 119: gy. J Allergy Clin Immunol 1997; 99: 179–
Cunha FQ, Silva TA, Nicoli JR, Vieira LQ, 305–309. 185.
Souza DG, Teixeira MM: Commensal mi- 81 Gionchetti P, Rizzello F, Helwig U, Venturi 89 Abrahamsson TR, Jakobsson T, Bottcher
crobiota is fundamental for the development A, Lammers KM, Brigidi P, Vitali B, Poggio- MF, Fredrikson M, Jenmalm MC, Bjorksten
of inflammatory pain. Proc Natl Acad Sci li G, Miglioli M, Campieri M: Prophylaxis of B, Oldaeus G: Probiotics in prevention of
USA 2008;105:2193–2197. pouchitis onset with probiotic therapy: a IgE-associated eczema: a double-blind, ran-
74 Bravo JA, Forsythe P, Chew MV, Escaravage double-blind, placebo-controlled trial. Gas- domized, placebo-controlled trial. J Allergy
E, Savignac HM, Dinan TG, Bienenstock J, troenterology 2003;124:1202–1209. Clin Immunol 2007;119:1174–1180.
Cryan JF: Ingestion of Lactobacillus strain 82 Whelan K, Quigley EM: Probiotics in the 90 Fiocchi A, Burks W, Bahna SL, Bielory L,
regulates emotional behavior and central management of irritable bowel syndrome Boyle RJ, Cocco R, Dreborg S, Goodman R,
GABA receptor expression in a mouse via the and inflammatory bowel disease. Curr Opin Kuitunen M, Haahtela T, Heine RG, Lack G,
vagus nerve. Proc Natl Acad Sci USA 2011; Gastroenterol 2013; 29:184–189. Osborn DA, Sampson H, Tannock GW, Lee
108:16050–16055. 83 Papa E, Docktor M, Smillie C, Weber S, Pre- BW: Clinical use of probiotics in pediatric al-
75 Bercik P, Denou E, Collins J, Jackson W, Lu J, heim SP, Gevers D, Giannoukos G, Ciulla D, lergy (CUPPA): a World Allergy Organiza-
Jury J, Deng Y, Blennerhassett P, Macri J, Mc- Tabbaa D, Ingram J, Schauer DB, Ward DV, tion position paper. World Allergy Organ J
Coy KD, Verdu EF, Collins SM: The intestinal Korzenik JR, Xavier RJ, Bousvaros A, Alm 2012;5:148–167.
microbiota affect central levels of brain-de- EJ: Non-invasive mapping of the gastrointes- 91 Hilty M, Burke C, Pedro H, Cardenas P, Bush
rived neurotropic factor and behavior in mice. tinal microbiota identifies children with in- A, Bossley C, Davies J, Ervine A, Poulter L,
Gastroenterology 2011;141:599–609, e1–e3. flammatory bowel disease. PLoS One 2012; Pachter L, Moffatt MF, Cookson WO: Disor-
76 Tillisch K, Labus J, Kilpatrick L, Jiang Z, 7:e39242. dered microbial communities in asthmatic
Stains J, Ebrat B, Guyonnet D, Legrain-Ras- 84 Gao Z, Perez-Perez GI, Chen Y, Blaser MJ: airways. PLoS One 2010;5:e8578.
paud S, Trotin B, Naliboff B, Mayer EA: Con- Quantitation of major human cutaneous 92 Cardenas PA, Cooper PJ, Cox MJ, Chico M,
sumption of fermented milk product with bacterial and fungal populations. J Clin Mi- Arias C, Moffatt MF, Cookson WO: Upper
probiotic modulates brain activity. Gastro- crobiol 2010;48:3575–3581. airways microbiota in antibiotic-naive
enterology 2013; 144:1394–1401.e4. 85 Findley K, Oh J, Yang J, Conlan S, Deming C, wheezing and healthy infants from the trop-
77 Frank DN, St Amand AL, Feldman RA, Meyer JA, Schoenfeld D, Nomicos E, Park M, ics of rural Ecuador. PLoS One 2012;
Boedeker EC, Harpaz N, Pace NR: Molecu- Becker J, Benjamin B, Blakesley R, Bouffard 7:e46803.
lar-phylogenetic characterization of micro- G, Brooks S, Coleman H, Dekhtyar M, Greg- 93 Thomas CM, Versalovic J: Probiotics-host
bial community imbalances in human in- ory M, Guan X, Gupta J, Han J, Hargrove A, communication: modulation of signaling
flammatory bowel diseases. Proc Natl Acad Ho SL, Johnson T, Legaspi R, Lovett S, Ma- pathways in the intestine. Gut Microbes
Sci USA 2007;104:13780–13785. duro Q, Masiello C, Maskeri B, McDowell J, 2010;1:148–163.

Universidade Federal do Rio de Janeiro


146.164.3.22 - 1/27/2014 4:15:39 AM

52 Ann Nutr Metab 2013;63(suppl 2):42–52 Versalovic


DOI: 10.1159/000354899
Downloaded by:

You might also like