<<

Pharmacological Research 69 (2013) 1–10

Contents lists available at SciVerse ScienceDirect

Pharmacological Research

jo urnal homepage: www.elsevier.com/locate/yphrs

Invited Review

The human milk microbiota: Origin and potential roles in health and disease

a a,b a a,c

Leónides Fernández , Susana Langa , Virginia Martín , Antonio Maldonado ,

a d a,∗

Esther Jiménez , Rocío Martín , Juan M. Rodríguez

a

Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain

b

Departament of Food Technology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Carretera de la Coru˜na Km 7.5, 28040 Madrid, Spain

c

Departament of Food Biotechnology, Instituto de la Grasa-CSIC, 41012 Sevilla, Spain

d

Danone Research-Centre for Specialised Nutrition, Wageningen, The Netherlands

a r t i c l e i n f o a b s t r a c t

Article history: Human milk has been traditionally considered sterile; however, recent studies have shown that it repre-

Received 22 May 2012

sents a continuous supply of commensal, mutualistic and/or potentially to the infant

Received in revised form 23 August 2012

gut. Culture-dependent and -independent techniques have revealed the dominance of staphylococci,

Accepted 1 September 2012

streptococci, bacteria and bifidobacteria in this biological fluid, and their role on the coloniza-

tion of the infant gut. These bacteria could protect the infant against infections and contribute to the

Keywords:

maturation of the immune system, among other functions. Different studies suggest that some bacteria

Human milk

present in the maternal gut could reach the mammary gland during late pregnancy and lactation through

Breastfeeding

Bacteria a mechanism involving gut monocytes. Thus, modulation of maternal during pregnancy

and lactation could have a direct effect on infant health. On the other hand, mammary dysbiosis may lead

Mammary microbiota

Dendritic cells to mastitis, a condition that represents the first medical cause for undesired weaning. Selected strains

Mastitis isolated from breast milk can be good candidates for use as . In this review, their potential uses

Probiotics for the treatment of mastitis and to inhibit mother-to-infant transfer of HIV are discussed. HIV © 2012 Elsevier Ltd. All rights reserved.

Contents

1. Bacterial diversity of human milk ...... 1

2. Functions of human milk bacteria in the infant gut ...... 3

3. Origin of the bacteria isolated from breast milk: is there a bacterial entero-mammary pathway? ...... 3

4. Human milk bacteria as biotherapeutic agents ...... 5

4.1. Specific targets for human milk probiotic bacteria: lactational mastitis ...... 6

4.2. Specific targets for human milk probiotic bacteria: anti-HIV activity ...... 6

5. Conclusion...... 7

Acknowledgements ...... 7

References ...... 7

1. Bacterial diversity of human milk present in colostrum and milk, including immunocompetent

cells, immunoglobulins, fatty acids, polyamines, oligosaccharides,

Human milk is a complex -specific biological fluid lysozyme, lactoferrin and other glycoproteins, and

adapted to satisfy the nutritional requirements of the rapidly grow- peptides [2], which inactivate pathogens individually, additively,

ing infant; additionally, it educates the infant immune system and and synergistically [3].

confers a certain degree of protection against pathogens [1]. These More recently, several studies have revealed that colostrum and

effects reflect the synergistic action of many bioactive molecules, breast milk are continuous sources of commensal, mutualistic and

potentially probiotic bacteria to the infant gut [4–11] (Table 1). This

is a relevant finding since intramammary human milk was tradi-

tionally considered to be sterile. In fact, human milk constitutes

Abbreviations: DCs, dendritic cells; HIV, human immunodeficiency virus; LAB,

one of the main sources of bacteria to the breastfed infant gut since .

a baby consuming approximately 800 mL/day of milk would ingest

Corresponding author. Tel.: +34 91 3943837; fax: +34 91 3943743.

5 7

× ×

E-mail address: [email protected] (J.M. Rodríguez). between 1 10 and 1 10 bacteria daily [5]. This may explain

1043-6618/$ – see front matter © 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.phrs.2012.09.001

2 L. Fernández et al. / Pharmacological Research 69 (2013) 1–10

Table 1

showed the predominance of staphylococci, streptococci, lactic acid

Main bacterial genera or species isolated from human milk or which DNA sequencies

bacteria (LAB), propionibacteria and closely related Gram-positive

have been retrieved from this biological fluid.

bacteria [4,5,18,19] (Table 1), including new bacterial species, such

a

Method Main species/genera References

as lactarius [20]. Human milk has also been shown

Bacterial isolation L. acidophilus, L. fermentum, S. epidermidis, [18] to be a source of live bifidobacteria to the infant gut [11]. The fact

Str. mitis, Str. salivarius that bacteria belonging to such genera can be isolated from fresh

L. plantarum, S. epidermidis, Streptococcus [19]

breast milk of healthy women from distant countries suggests that

spp.

their presence in this substrate is a common event. Therefore, they

E. faecium, L. fermentum, L. gasseri [4]

should be considered as components of the natural microbiota of

E. faecalis, L. crispatus, L. rhamnosus, Lc. [5,6]

lactis, Leuc. mesenteroides, R. mucilaginosa, human mammary gland, instead of mere contaminant bacteria. The

S. aureus, S. capitis, S. epidermidis, S.

mammary microbiota is somehow peculiar since it has a transient

hominis, Str. mitis, Str. oris, Str. parasanguis,

nature: its development starts during the last third of pregnancy,

Str. salivarius

reaches the highest complexity at the end of such period, remains

L. salivarius [10]

Corynebacterium spp., Enterococcus spp., [63] quite constant throughout lactation, declines sharply at weaning,

Lactobacillus spp., Peptostreptococcus spp., and rapidly disappears when there is no milk in the mammary gland

Staphylococcus spp., Streptococcus spp.

and the apoptosis process responsible for mammary involution

L. reuteri [120]

begins (Fig. 1).

S. epidermidis [7,8]

Several studies have shown that there is a mother-to-infant

B. adolescentis, B. bifidum, B. breve [11]

B. breve, B. longum, K. rhizophila, L. casei, L. [20,24] transfer of bacterial strains belonging, at least, to the genera Lacto-

fermentum, L. gasseri, L. gastricus, L.

bacillus, , Enterococcus, and Bifidobacterium through

plantarum, L. reuteri, L. salivarius, L.

breastfeeding [4,7,11,21–24]. Independently of the original source

vaginalis, P. pentosaceus, R. mucilaginosa, S.

of such bacteria, human milk is the source of hundreds of bacterial

aureus, S. epidermidis, S. hominis, Str.

lactarius, Str. mitis, Str. parasanguis, Str. phylotypes to the infant . It has been suggested

salivarius that exposure of the breast-fed infant to such a wealth of bacte-

E. durans, E. faecalis, E. faecium, E. hirae, E. [22]

rial phylotypes may exert beneficial effects against diarrheal and

mundtii, L. animalis, L. brevis, L. fermentum,

respiratory diseases, and may reduce the risk of developing other

L. gasseri, L. helveticus, L. oris, L. plantarum,

diseases, such as diabetes or obesity [25,26].

P. pentosaceous, Str. australis, Str.

gallolyticus, Str. vestibularis The application of culture-independent molecular techniques,

B. longum [23]

and particularly those based on 16S rRNA genes, allowed a

complementary biodiversity assessment of the human milk micro-

DNA detection E. faecalis, E. faecium, L. fermentum, L. [27,28]

gasseri, L. rhamnosus, Lc. lactis, Leuc. biome. The use of such techniques confirmed the dominance of

citreum, Leuc. fallax, Prop. acnes, S.

staphylococci and streptococci and the presence of LAB, propi-

epidermidis, S. hominis, Str. mitis, Str.

onibacteria and bifidobacteria, and revealed the existence of DNA

parasanguis, Str. salivarius, W. cibaria, W.

confusa belonging to other bacterial groups, such as some Gram-negative

B. longum, Clostridium spp., [65] bacteria [11,27–29]. In addition, the application of the “-omics”

spp., Staphylococcus spp., Streptococcus spp. approach (genomics, metagenomics, transcriptomics, proteomics,

B. adolescentis B. animalis, B. bifidum, B. [29]

and metabolomics) to the study of the human mammary micro-

breve, B. catenolatum, B. longum

biota is already in progress and there is no doubt that the results

Bifidobacterium spp., Clostridium spp., [121]

provided by such techniques will open new perspectives to under-

Enterococcus spp., Lactobacillus spp.,

Staphylococcus spp., Streptococcus spp. stand the initiation and development of the infant gut microbiota

B. adolescentis, B. bifidum, B. breve, B. [11] [30].

longum

Recently, the first microbiome study focused on human milk

Bradyrhizobiaceae, Corynebacterium spp., [25]

was published [25]. The authors used microbial identification

Propionibacterium spp., Pseudomonas spp.,

Ralstonia spp., Serratia spp., Sphingomonas techniques based on pyrosequencing of the V1–V2 region of the

spp., Staphylococcus spp., Streptococcus spp. bacterial 16S rRNA gene to characterize the bacterial communities

a present in milk samples collected from 16 women self-described as

Abbreviations: B., Bifidobacterium; E., Enterococcus; K., Kocuria; L., Lactobacil-

lus; Lc., Lactococcus; Leuc., Leuconostoc; P., Pediococcus; Prop., Propionibacterium; R., healthy at three time-points over 4 weeks. Results indicated that

Rothia; S., Staphylococcus; Str., Streptococcus; W., Weissella.

milk bacterial communities were generally complex and, although

a few genera (Streptococcus, Staphylococcus, and Serratia) repre-

sented greater than 5% of the relative community abundance,

why the bacterial composition of the gut microbiota of breast- eight other genera represented ≥1% of the communities observed

fed infants is closely related to that found in the breast milk of across samples. Among the hundreds of operational taxonomic

their respective mothers and, also, why the development of a more units (OTUs) detected in the milk of every woman, only 9 (Strep-

diverse microbiota coincides with the weaning period [12]. It must tococcus, Staphylococcus, Serratia, Pseudomonas, Corynebacteria,

be highlighted that human milk olisaccharides (HMOs) also play a Ralstonia, Propionibacterium, Sphingomonas, and Bradyrhizobiaceae)

key role in driving the diversity of the infant gut microbiota [13]. were present in every sample from every woman. These 9 “core”

HMOs are the third-largest solid component of milk and, while their OTUs represented approximately half of the microbial commu-

structural complexity renders them non-digestible to the host, they nity observed, although their relative abundance varied greatly

are liable to hydrolytic enzymes of the infant colonic microbiota between subjects. The remaining half of the community was not

[14]. It has been previously demonstrated that selected bifidobac- conserved across women. These findings are in contrast with those

terial and lactobacilli phylotypes use specific HMOs secreted early of the gut microbiome, where only a low set of OTUs is shared

in the lactation cycle [15]; more recently, the genome sequences among individuals [31], or the vaginal microbiome which com-

of two bifidobacterial strains have revealed several adaptations for prises several different core groups [32]. On the other hand, milk

milk utilization within the infant microbiome [16,17]. bacterial community was generally stable over time within an indi-

The first descriptions of the bacterial diversity of breast milk vidual. Similarly to the mammary microbiota, previous studies of

in healthy women were based on the use of culture media and various sites of the human microbiome have also revealed that the

L. Fernández et al. / Pharmacological Research 69 (2013) 1–10 3

• Hormonal signalling • Increasing recruitment of • Sharpe decrease in milk

103 DCs and macrophages production • Increasing pressure of • Decreasing the fetus on the mesenteric concentrations of lactose

vessels in the mammary • Increased bacterial environment

translocation •Apoptosis • Increasing filling of the (physiological involution mammary ducts with of the mammary gland) colostrum Concentration of bacteria (CFU/ml)

Not detectable

Last third of pregnancy Peripartum Breastfeeding Weaning (~3 months) (a fewdays) (a few days-a few years) (some days-some weeks)

Time

Fig. 1. Schematic representation of the acquisition and development of the human mammary microbiota.

bacterial communities associated with a particular individual over However, both strains led to a reduction of Th1 cytokines when cells

time are often stable and highly personalized [33]. were incubated in the presence of lipopolysaccharide [40]. A recent

The results of a second study on the breast milk microbiome study confirmed that L. fermentum CECT 5716 and L. salivarius CECT

involving 18 mothers indicated that milk bacteria are not contami- 5713 have a broad array of effects on the immune system [43]. They

nants and suggested that this site-specific microbiome is influenced behaved as potent activators of NK cells and moderate activators

by several factors that significantly skew its composition [34]. More of CD4+ and CD8+ T cells and regulatory T cells. Thus, they had

recently, our research group analyzed the metagenome of human an impact on both innate and acquired immunity. They strongly

milk samples obtained from healthy, mastitis-suffering or obese induced a wide range of pro- and anti-inflammatory cytokines

women, and observed that the bacterial community of breast milk and chemokines. The authors compared these strains with others

may differ depending on the individual and on the health status of belonging to the same species but isolated from sources different

the lactating women [unpublished results]. to breast milk and found some milk strain-specific effects, such as

a higher induction of IL-10 and IL-1 production.

In relation to allergic conditions, the human milk strain Lacto-

2. Functions of human milk bacteria in the infant gut

bacillus gasseri CECT 5714 reduced the incidence and severity of the

allergic response in an animal model of cow’s milk protein allergy

In the last years, some studies have shown that human milk

[44]. It is also interesting to note that the presence of viridans strep-

bacteria may play several roles in the infant gut. First of all,

tococci, one of the dominant bacterial groups in human milk, seems

they can contribute to the reduction of the incidence and sever-

to be a feature of the healthy infant gut in contrast to that of the

ity of infections in the breastfed infant by different mechanisms,

atopy-suffering infants [45].

such as competitive exclusion [35], production of antimicrobial

Finally, human milk bacteria have a remarkable potential to play

compounds [6,9,10,35], or improvement of the intestinal barrier

metabolic roles in the infant. The glycobiome of some lactobacilli

function by increasing mucine production and reducing intestinal

and bifidobacteria, including that of species that have been isolated

permeability [35]. Recently, the administration of a human milk

from human milk, may help to create a specific “healthy” micro-

Lactobacillus strain to infants during 6 months led to 46%, 27%, and

biota in the infant gut [14,46]. These might also

30% reductions in the incidence rates of gastrointestinal infections,

contribute to infant digestion through the breakdown of sugars and

upper respiratory tract infections, and total number of infections,

proteins; this possibility is particularly attractive having in account

respectively [36]. Commensal coagulase-negative staphylococci

that transit of food through the gastrointestinal tract is shorter in

and viridans streptococci provided by breast milk can be partic-

infants than in adults and, also, that the pH of the infant’s stomach

ularly useful to reduce the acquisition of undesired pathogens by

is higher than that of the adult. In this context, human milk lacto-

infants exposed to hospital environments. In fact, some Staphy-

strains are metabolically active in the infant gut and increase

lococcus epidermidis strains that inhibit in vivo colonization by

the production of functional metabolites such as butyrate, which is

Staphylococcus aureus have been postulated as a future strategy to

the main energy source for colonocytes and a relevant compound

eradicate such pathogen from the mucosal surfaces [37,38]. Sim-

in the modulation of intestinal function [47,48]. As a result, they

ilarly, it has been shown that viridans streptococci inhibit oral

improve the intestinal habit, with an increase in fecal moisture,

colonization by methicillin-resistant S. aureus in high-risk new-

and in stool frequency and volume [47,48].

borns exposed to hospital environments [39].

Breast milk bacteria may also participate in the correct mat-

uration of the infant immune system since some strains are able 3. Origin of the bacteria isolated from breast milk: is there

to modulate both natural and acquired immune responses in mice a bacterial entero-mammary pathway?

and humans [40–42]. Their function seems to have a certain degree

of flexibility depending on the conditions found in the gut envi- The origin of the bacteria present in breast milk has become a

ronment. As an example, Lactobacillus salivarius CECT 5713 and controversial issue in the last years. Traditionally, it was believed

Lactobacillus fermentum CECT 5716 enhanced macrophage produc- that milk harbored bacteria that were just the result of contamina-

tion of Th1 cytokines, such as IL-2 and IL-12 and the inflammatory tion with bacteria from the mother’s skin or the infant’s oral cavity

mediator TNF-␣, in the absence of an inflammatory stimulus. [19].

4 L. Fernández et al. / Pharmacological Research 69 (2013) 1–10

Fig. 2. Potential sources of the bacteria present in human colostrum and milk. DC: dendritic cell.

Infrared photography [49] has shown that a certain degree of least, a very unlikely source [29]. These and other findings suggest

retrograde flow back into the mammary ducts can occur during that at least some of the bacteria present in the maternal gut could

suckling. Obviously, such back flow may provide an ideal route for reach the mammary gland through an endogenous route [56].

the exchange of bacteria from the infant’s mouth into the mam- Although the pathway and mechanisms that some bacteria

mary gland (Fig. 2). But it is also obvious that breast milk can also could exploit to cross the intestinal epithelium and reach the mam-

be a source of bacteria to the infant’s mouth. Ecological niches in the mary gland and other locations has not been elucidated yet, some

human microbiome are not thought to be isolated environments, works have offered a plausible scientific basis. It has been demon-

but rather a network of interrelated communities experiencing strated that dendritic cells (DCs) can penetrate the gut epithelium

constant exchange [33]. Therefore, it is very likely that milk or to take up non-pathogenic bacteria directly from the gut lumen.

mammary bacterial communities are no the exception, and that DCs are able to open the tight junctions between intestinal epithe-

they are constantly influenced by exposure to other microbial pop- lial cells, send dendrites outside the epithelium and directly sample

ulations associated with the mother and her infant. Little is known bacteria, while preserving the integrity of the epithelial barrier

about the infant human salivary microbiome but investigations on through the expression of tight-junction proteins [57]. Using such

adults have revealed that Streptococcus species are the dominant mechanism, a typhimurium strain that was deficient in

phylotype in this fluid [50–52] while such dominance is even higher invasion genes encoded by Salmonella pathogenicity island 1 (SPI1)

in edentulous infants [53]. Streptococcus species are among the was still able to reach the spleen after oral administration to mice

most abundant phylotype in colostrum and milk samples [7,8,25], [57]. This mechanism may not be exclusive to DCs, as CD18+ cells,

supporting the hypothesis that the maternal milk bacteria may play including macrophages, have been shown to be essential for extra-

an important role in establishing salivary bacterial communities intestinal dissemination of non-invasive Salmonella [58].

and/or vice versa. It has also been shown that intestinal DCs can retain small num-

Some bacterial phylotypes usually present on adult skin, such bers of live commensal bacteria for several days in the mesenteric

as Staphylococcus, Corynebacteria, and Propionibacteria [54,55], are lymph nodes [59]. Once inside DCs and/or macrophages, gut bac-

also common in human milk. This presents the possibility that teria could spread to other locations since there is a circulation

interactions with the maternal skin microbiota may also contribute of lymphocytes within the mucosal associated lymphoid system.

to shape the composition of milk microbiota (Fig. 2). However, a Antigen-stimulated cells move from the intestinal mucosa to colo-

comparison of the bacterial communities detected in milk to those nize distant mucosal surfaces, such as those of the respiratory and

of the sebaceous skin found on the breast indicates that although genitourinary tracts, salivary and lachrymal glands, and, most sig-

the two communities share some phylotypes, major differences nificantly, that of the lactating mammary gland [60]. In addition, it is

also exist [25]. Such relevant differences between the two commu- known that, during the lactation period, colonization of the mam-

nities indicate that bacterial communities in milk are not simply mary gland by cells of the immune system is a selective process

a result of skin contamination. Sampling of breast milk for micro- regulated by the lactogenic hormones [61]. This process is respon-

biological analysis must take into account that skin contamination sible for the abundance of such cells in human milk. A hypothetical

is almost unavoidable and that, logically, doubts on the original model to explain how some maternal bacteria could be transferred

location (internal mammary gland or skin) of the isolated bacteria to the neonatal gut is shown in Fig. 2.

may arise. However, independent studies have shown that lacto- The suggestion that the origin of the live bacteria found in breast

bacilli or bifidobacteria could not be isolated from breast skin swabs milk could be the maternal gut and that the bacteria would arrive

obtained from women that provided milk samples from which such to the mammary gland through an endogenous route, involving

bacteria were isolated [11,29]. Previously, it has been reported that maternal DCs and macrophages, has been confirmed recently by

lactobacilli and enterococcal isolates present in human milk are independent research groups. Initially, our group showed that the

genotypically different from those isolated in the skin, within a exposure of mouse immature DCs to two bacterial strains isolated

same bacterial species and a same host [4]. In addition, bifidobac- from human milk led to a high stimulation of two DC activation

teria belongs to a strictly anaerobic genus and, therefore, skin is, at surface markers: the class II major histocompatibility complex and

L. Fernández et al. / Pharmacological Research 69 (2013) 1–10 5

At the same time, the whole body (including immune, cardiovascu-

lar, respiratory, digestive and genitourinary systems) experiences

diverse physiological adaptations during pregnancy and lactation,

and most of them are compatible with an increase in the bacterial

translocation rate in the gut.

It has to be highlighted that the presence of live LAB (similar to

those found in human milk) in the bloodstream of healthy human

hosts is not an uncommon event [69–72]. It is interesting to note

that 125 of the 485 lactobacilli strains deposited in the PROSAFE

collection were originally isolated from human blood, and it is

also illustrative that these 125 strains belong to 16 different Lacto-

bacillus species [69]. A study investigating the influence of the oral

microbiota composition on pregnancy outcome in 300 pregnant

women revealed that some bacteria, such as Actinomyces naselundii,

were linked to lower birth weight and earlier delivery while oth-

ers, including , were associated with a slightly

higher birth weight and normal delivery [73]. These authors con-

cluded that oral bacteria can enter the uterine environment through

the bloodstream. Probably, in addition to the digestive and geni-

tourinary tracts, LAB and other bacteria can inhabit, permanent- or

Fig. 3. Specific interactions between cells of a L. gasseri strain isolated from breast

transiently, other body locations in healthy hosts, where they could

milk (A) and DC cells (B), as assessed by transmission electron microscopy [62].

contribute to extraintestinal probiotic effects. Additionally, human

The interactions were studied using transwell bicompartmental assays in which

bacterial cells and immature DC cells were initially separated by a monolayer of clinical studies have also indicated that oral intake of specific bac-

Caco-2 cells. terial strains at defined doses play a key role in the prevention

or improvement of extraintestinal conditions, and such probiotic

the B7.2 protein [62,63]. It seems clear that bacterial induction of effects would be hardly explainable if they were exclusively con-

DC maturation is an active process since it has been shown that fined to the gut [74]. Globally, these studies indicate that certain

dead bacteria or inert particles, such as latex beads, cannot activate intestinal bacteria may have a rather underrated ability to spread

DCs even if they are rapidly phagocytosed [64]. On the other hand, L. from the gut to extra-intestinal locations in healthy hosts.

gasseri CECT 5715, other strain isolated from breast milk, showed a

high level of binding to DCs and ability to translocate across a Caco-2

cell monolayer through a DC-mediated mechanism [62] (Fig. 3). 4. Human milk bacteria as biotherapeutic agents

Later, other study showed that bacterial translocation from the

gut to mesenteric lymph nodes and mammary gland occurred dur- In recent years, the problems associated to the spread of clin-

ing late pregnancy and lactation in mice [65]. In addition, this ical resistances among pathogenic bacteria and to the

work revealed that human breast milk cells contain a number of rise of allergic or inflammatory diseases in developed countries

viable bacteria and a range of bacterial DNA signatures, which are have led to a new interest in probiotics, which are defined as live

also found in maternal peripheral blood mononuclear cells. Those microorganisms that confer a health benefit on the host when

peripheral blood mononuclear cells showed greater biodiversity administered in adequate amounts [75]. Obviously, probiotic bac-

than did those obtained from control women. Taken together, teria that are originally isolated from human milk are particularly

their results suggest that intestinally derived bacteria and bac- attractive organisms since they would fulfill some of the main crite-

terial components are transported to the lactating breast within ria generally recommended for human probiotics, such as human

mononuclear cells. The authors speculated that this programs the origin, a history of safe prolonged intake by a particularly sensi-

neonatal immune system to recognize specific bacterial molecular tive population (neonates, infants), and adaptation to mucosal and

patterns and to respond appropriately to pathogenic and com- dairy substrates [76].

mensal organisms [65]. More recently, two successive studies that Among the bacteria isolated from human milk, species like L.

focused on the oral administration of three lactobacilli strains iso- gasseri, L. salivarius, Lactobacillus reuteri, L. fermentum or Bifidobac-

lated from human milk (L. salivarius CECT 5713, L. gasseri CECT 5714, terium breve are considered among those with probiotic potential

and L. fermentum CECT 5716) provided new evidences that show the and enjoy the Qualified Presumption of Safety (QPS) status con-

existence of a bacterial entero-mammary pathway during lactation ceded by the European Food Safety Authority (EFSA). In contrast

[66,67]. to other bacteria, these seem to be uniquely adapted to reside

The mammary gland prepares for lactation through a series of in the human digestive tract and to interact with us in symbio-

developmental steps that occur during adolescence and pregnancy. sis from the time we are born. Additionally, some of the strains

The principal feature of mammary growth in pregnancy is a great isolated from this biological fluid have been shown to play anti-

increase in ducts and alveoli under a multi-hormonal influence. At infectious, anti-inflammatory, immunomodulatory and metabolic

the end of this period, the lobules of the alveolar system are maxi- roles, both in vitro and in vivo, including human studies (see Sec-

mally developed and small amounts of colostrum may be released tion 2). The recent genome sequencing of some of these strains

for several weeks prior to delivery. Additionally, the nipple and [77–80] is providing some new clues to understand the relation-

areola markedly enlarge and the sebaceous glands within become ship between phenotypic properties and their subjacent molecular

more prominent [68]. The increased lymph and blood supply to basis [63]; in the future, such approach may contribute to a more

the mammary gland and the oxytocin release that causes contrac- rational selection, design and/or application of probiotic bacteria.

tion of the mioepithelial cells that invest the mammary alveoli may Exclusive breastfeeding during the first month of life has been

also facilitate the presence of endogenous bacteria in breast milk. linked to lower asthma [81] and atopic dermatitis [82] rates during

These changes provide good conditions for biofilm formation on childhood and, as a result, this practice has been strongly recom-

the mammary areola and/or in the mammary duct system, leading mended to mothers with a family of atopy, as a possible means

to the formation of a specific and transitory mammary microbiota. to prevent atopic eczema. Human milk LAB may play a role in

6 L. Fernández et al. / Pharmacological Research 69 (2013) 1–10

this protective effect since it has been described that probiotic respectively. In contrast, the effectiveness of prescribed

lactobacilli can be effective to prevent atopy and atopic diseases to group C women differed significantly, both in the reduction of

through a variety of mechanisms [83]. Gut bacteria are considered bacterial counts and in the improvement of the pain score.

the earliest and most important stimulus for development of gut-

associated lymphoid tissue and they can promote antiallergenic 4.2. Specific targets for human milk probiotic bacteria: anti-HIV

processes [83]. Therefore, milk of healthy women can be consid- activity

ered as a source of potentially probiotic or biotherapeutic bacteria

with a role in protecting mothers and/or infants against a variety Although breastfeeding is a source of new pediatric HIV-1 infec-

of allergic, inflammatory or infectious diseases. tions worldwide [96,97], most breastfed infants of HIV-positive

women remain uninfected despite repeated exposure of their oral

4.1. Specific targets for human milk probiotic bacteria: and gastrointestinal mucosal surfaces to the virus. Therefore, breast

lactational mastitis milk has been recognized both as a vector of transmission and as

vehicle of protection against HIV-1 [96].

Mastitis is a common disease during lactation since it has an Most breast milk-transmitted HIV infections take place in devel-

incidence of up to 33% of the lactating mothers [84,85]. This inflam- oping countries where breastfeeding usually remains the best

mation of one or more lobules of the mammary gland usually has an election for providing optimal infant nutrition and protection

infectious origin [86], involving staphylococci, streptococci and/or against morbidity and mortality associated with diarrheal and

corynebacteria [85]. Traditionally, S. aureus has been considered as lower respiratory infections [98,99]. In such countries, the choice

the main etiological agent of acute mastitis although S. epidermidis of the most suitable infant feeding option for an HIV-infected

is emerging as the leading cause of subacute and chronic mastitis mother should depend on her individual circumstances, including

both in human and veterinary medicine [87–90]. Multiresistance to her health status and the local situation, and should also consider

antibiotics, formation of biofilms and mechanisms for evasion of the the health services available and the counseling and support she is

host immune response are common among clinical isolates of these likely to receive [100].

two staphylococcal species [91–95]. This explains why this condi- As a general rule, exclusive breastfeeding has been recom-

tion uses to be elusive to antibiotic therapy, and why it constitutes mended for HIV-infected women during the first 6 months of

one of the main reasons to cease breastfeeding [85]. In this con- life unless replacement feeding is acceptable, feasible, affordable,

text, the development of new strategies for mastitis management sustainable and safe [97,100]. Such feeding option may offer HIV-

based on probiotics, as an alternative or complement to antibiotic 1-infected women in developing countries an affordable, culturally

therapy, is particularly appealing. acceptable, and effective means of reducing mother-to-child HIV-

For this purpose, our research group selected some lactobacilli 1 transmission while maintaining the benefits of breastfeeding

strains on the basis of specific properties required for success in [101]. In fact, recent studies confirm that exclusive breastfeeding

mastitis treatment after oral administration: a high survival rate can be successfully supported in HIV-infected women [102,103]. In

during transit through the gastrointestinal tract, specific interac- contrast, there seems to be an association between mixed breast-

tions with DCs, ability to colonize the mammary gland and, once feeding and increased HIV transmission risk [104].

there, mechanisms for competitive exclusion of mastitis-causing Several factors may modulate the risk of HIV-1 transmission

staphylococci and streptococci. Initially, a pilot trial highlighted via breastfeeding but the results of different studies focused on

the potential of L. salivarius CECT 5713 and L. gasseri CECT 5714, the potential role of specific milk components are not conclusive

two strains isolated from breast milk, for the treatment of staphy- [105–107]. Recently, it was shown that breast milk contains innate

lococcal mastitis [66]. After 30 days, probiotics led to a significant factors that blocked infection of CD4+ cells with cell-free HIV, but

reduction ( 2 log10 CFU/mL) in the mean staphylococcal counts of did not affect infection with cell-associated virus [107]. On the other

the milk cultures, in contrast, the values achieved in the placebo hand, humoral mucosal immunity to HIV does not appear to be

group remained unchanged at the end of the study. This fact was a protective factor against viral transmission through breast milk

correlated with the clinical evolution since, while no clinical signs [105,108]. Therefore, factors associated to protection against ver-

of mastitis were observed in women assigned to the probiotic group tical transmission of HIV-1 through breastfeeding remain poorly

at day 14, they persisted throughout the study in the placebo group. understood [96].

More recently, the efficacy of L. fermentum CECT 5716 or L. Interestingly, the potential of some human milk strains to inhibit

salivarius CECT 5713, two lactobacilli strains isolated from breast the infectivity of HIV with tropism to different co-receptors has

milk, to treat lactational mastitis when administered orally was been described recently [109]. A total of 38 bacterial strains iso-

evaluated and compared to antibiotic therapy [67]. A total of lated from breast milk were investigated for their ability to inhibit

352 women with infectious mastitis were randomly divided HIV-1 infection in vitro. The heat-killed bacteria and cell-free super-

in three groups. Those in groups A (n = 124) and B (n = 127) natants obtained from some strains inhibited HIV-1 infection; the

ingested daily 9 log10 CFU of L. fermentum CECT 5716 or L. sali- highest levels corresponded to killed bacteria from Lactobacillus

varius CECT 5713, respectively, for 3 weeks while those in group curvatus VM25 (55.5%), L. fermentum VM31 (52.5%), Pediococcus

C (n = 101) were submitted to antibiotic therapy prescribed in pentosaceous VM95 (49.0%), and P. pentosaceous VM21 (45.5%).

their respective Primary Care Centres. On day 0, the mean bac- HIV-inhibition was observed with supernatants from eight strains,

terial counts in milk samples of the three groups were similar although the most effective were L. salivarius VM5 (42%) and L.

(4.35–4.47 log10 CFU/mL) and lactobacilli could not be detected. gasseri VM22 (40%) supernatants. Four strains (L. salivarius VM5, L.

On day 21, the mean bacterial counts in the probiotic groups (2.61 gasseri VM22, VM17, and Streptococcus salivarius

and 2.33 log10 CFU/mL) were lower than that of the control group VM18) were selected for further evaluation and demonstrated dis-

(3.28 log10 CFU/mL). The probiotic treatment led to a significant tinct inhibition patterns against R5-, X4- and R5/X4-tropic HIV-1.

reduction (1.7–2.1 log10 CFU/mL) in the milk bacterial count and These results demonstrate for the first time that LAB from human

to a rapid improvement of the condition. The final bacterial count breast milk can significantly inhibit HIV-1 infection in vitro, and

was approximately 2.5 log10 CFU/mL, an acceptable bacterial load suggest a possible role for these bacteria in mucosal protection

in milk of healthy women [5,64]. After the probiotic treatment, L. against HIV-1 in breastfeed infants.

salivarius CECT 5713 and L. fermentum CECT 5716 could be isolated Human milk bacteria may exert an anti-HIV action in the

from the milk samples of women of the probiotic groups A and B, infant gut through a variety of mechanisms (Fig. 4), including

L. Fernández et al. / Pharmacological Research 69 (2013) 1–10 7

Fig. 4. Potential mechanisms by which human milk bacteria can inhibit HIV infectivity. (A) Interactions between DC, HIV, and bacteria: (A1) blocking of DC receptor through

proteins or exopolysaccharides and (A2) blocking of the gp120 glycoprotein from binding to target cells. (B) Potential effects of human milk bacteria at the mucosal sites.

interactions with and modulation of DCs immunological functions in gut microbiota and intestinal inflammatory parameters [117].

[110–112]. DC–HIV interaction is critical for the infection out- These findings support the hypothesis that gastrointestinal tract

come. Although DCs are able to induce specific immune responses alterations are a key factor in HIV pathogenesis, and may explain

against a broad variety of pathogens, HIV-1 has evolved ways to why infants who are exclusively breastfed have a significantly

exploit DCs as cellular reservoirs, therefore facilitating viral dis- lower risk of being HIV-infected than infants who are bottle-fed

semination, persistence in lymphoid tissues and evasion of antiviral or mixed-fed [118,119].

immunity [113,114]. Additionally, oral lactobacilli isolated from

healthy humans can capture HIV-1 in vitro through lectin-like

5. Conclusion

interactions between mannose residues on the viral envelope gly-

coprotein gp120 and the bacteria [115]. This physical interaction

Human milk is a source of bacteria to the infant gut, where

(the so-called “HIV-1 trap”) may prevent viral attaching to cellular

they may play a variety of antiinfectious, immunomodulatory, and

receptors on mucosal tissues.

metabolic roles. In fact, recent studies indicate that the mam-

Recently, it was observed that the genome of L. salivarius CECT

mary gland contains its own microbiota during late pregnancy and

5713 contains genes encoding four proteins (1184, 1230, 1306, and

lactation. This bacterial community may differ depending on the

1686) potentially involved in human molecular mimetism [63].

individual and the health status of the lactating women. It seems

Among them, protein 1230 was considered of particular relevance

that certain bacteria from the maternal gut can use mononuclear

because of its similarity with proteins related to DC functionality,

immune cells to colonize, first, the mammary gland and, later, the

such as CD209 antigen (DC-SIGN1) and the C-type lectin domain

infant gut through breast-feeding. If further studies confirm these

family 4 member M (DC-SIGN2). In addition, protein 1230 contains

findings, they would have practical consequences since it would

a recognition motif for high mannose N-linked oligosaccharides

imply that modulation of the maternal intestinal microbiota can

present in a variety of pathogen antigens, including HIV gp120.

have a direct effect on her infant’s health, opening new perspectives

Consequently, it might have the potential to block gp120 from bind-

for bacteriotherapy and probiotics.

ing to target cells and, therefore, to inhibit HIV infectivity. To test

such predictions, the ability of L. salivarius CECT 5713 to stimu-

Acknowledgements

late the maturation of immature DCs and to inhibit the in vitro

HIV-1 infectivity was assessed [63]. The heat-killed cells led to a

This study was partly supported by the FUN-C-FOOD

reduction in viral infectivity of TZM-bl target cells with R5 (HIV-

(Consolider-Ingenio 2010) and AGL2010-15420 projects from

1BaL), CXCR4 (HIV-1HC4) and R5/X4 (HIV-1C7/86) of 42.3, 58.9, and

the Ministerio de Economía y Competitividad (Spain).

49.8%, respectively (mean values). Although to a lesser extent, cell-

free conditioned supernatant from L. salivarius CECT 5713 cultures

also inhibited HIV-1BaL (23.5%), HIV-1HC4 (21.4%) and HIV-1C7/86 References

(17.6%) infectivity. The results of this work suggest that molecular

mimetism displayed by L. salivarius CECT 5713 may have an antag- [1] Morrow AL, Rangel JM. Human milk protection against infectious diarrhea:

implications for prevention and clinical care. Seminars in Pediatric Infectious

onist role toward HIV gp120 and, therefore, may explain, at least

Diseases 2004;15:221–8.

partly, its activity against HIV infectivity in vitro.

[2] Newburg DS. Innate immunity and human milk. Journal of Nutrition

Intestinal permeability decreases faster in breast-fed infants 2005;135:1308–12.

[3] Isaacs CE. Human milk inactivates pathogens individually, additively and syn-

than those given formula [116], suggesting that some components

ergistically. Journal of Nutrition 2005;135:1286–8.

in breast milk influence the maturation of the gut mucosal epithe-

[4] Martín R, Langa S, Reviriego C, Jiménez E, Marín ML, Xaus J, et al. Human

lium. Introduction of food proteins or enteric pathogens during milk is a source of lactic acid bacteria for the infant gut. Journal of Pediatrics

2003;143:754–8.

formula or mixed replacement feeding may increase the likeli-

[5] Heikkilä MP, Saris PEJ. Inhibition of Staphylococcus aureus by the commensal

hood of HIV transmission by stimulating intestinal inflammatory

bacteria of human milk. Journal of Applied Microbiology 2003;95:471–8.

responses in the neonatal gut, leading to increased permeability [6] Beasley SS, Saris PEJ. Nisin-producing Lactococcus lactis strains isolated from

human milk. Applied and Environment Microbiology 2004;70:5051–3.

and to facilitation of virus penetration. In fact, gastrointestinal tract

[7] Jiménez E, Delgado S, Maldonado A, Arroyo R, Albújar M, García N, et al.

impairment in HIV-positive patients is present in the early phases

Staphylococcus epidermidis: a differential trait of the fecal microbiota of

of HIV disease, and this impairment is associated with alterations breast-fed infants. BMC Microbiology 2008;8:143.

8 L. Fernández et al. / Pharmacological Research 69 (2013) 1–10

[8] Jiménez E, Fernández L, Delgado S, García N, Albújar M, Gómez A, et al. Academy of Sciences of the United States of America 2011;108(Suppl

Assessment of the bacterial diversity of human colostrum by cultural-based 1):4680–7.

techniques. Analysis of the staphylococcal and enterococcal populations. [33] Costello EK, Lauber CL, Hamady M, Fierer N, Gordon J, Knight R. Bacterial

Research in Microbiology 2008;159:595–601. community variation in human body habitats across space and time. Science

[9] Martín R, Olivares M, Marín ML, Fernández L, Xaus J, Rodríguez JM. Probiotic 2009;326:1694–7.

potential of 3 lactobacilli strains isolated from breast milk. Journal of Human [34] Cabrera-Rubio R, Collado MC, Laitinen K, Salminen S, Isolauri E, Mira A. The

Lactation 2005;21:8–17. human milk microbiome changes over lactation and is shaped by maternal

[10] Martín R, Jiménez E, Olivares M, Marín ML, Fernández L, Xaus J, et al. Lacto- weight and mode of delivery. American Journal of Clinical Nutrition 2012,

bacillus salivarius CECT 5713, a potential probiotic strain isolated from infant http://dx.doi.org/10.3945/ajcn.112.037382.

feces and breast milk of a mother–child pair. International Journal of Food

[35] Olivares M, Díaz-Ropero MP, Martín R, Rodríguez JM, Xaus J. Antimicrobial

Microbiology 2006;112:35–43.

potential of four Lactobacillus strains isolated from breast milk. Journal of

[11] Martín R, Jiménez E, Heilig HG, Fernández L, Marín ML, Zoetendal EG, et al.

Applied Microbiology 2006;101:72–9.

Isolation of bifidobacteria from breast milk and assessment of the bifi-

[36] Maldonado J, Canabate˜ F, Sempere L, Vela F, Sánchez AR, Narbona E,

dobacterial population by PCR-denaturing gradient gel electrophoresis and

et al. Human milk probiotic Lactobacillus fermentum CECT5716 reduces

quantitative real-time PCR. Applied and Environment Microbiology 2009;75:

the incidence of gastrointestinal and upper respiratory tract infections in

965–9.

infants. Journal of Pediatric Gastroenterology and Nutrition 2012;54(January

[12] Favier CF, Vaughan EE, de Vos WM, Akkermans ADL. Molecular monitor- (1)):55–61.

ing of succession of bacterial communities in human neonates. Applied and

[37] Iwase T, Uehara Y, Shinji H, Tajima A, Seo H, Takada K, et al. Staphylococcus

Environment Microbiology 2002;68:219–26.

epidermidis Esp inhibits Staphylococcus aureus biofilm formation and nasal

[13] Sela DA, Mills DA. Nursing our microbiota: molecular linkages between

colonization. Nature 2010;465:346–9.

bifidobacteria and milk oligosaccharides. Trends in Microbiology

[38] Park B, Iwase T, Liu GY. Intranasal application of S. epidermidis prevents col-

2010;18:298–307.

onization by methicillin-resistant Staphylococcus aureus in mice. PLoS ONE

[14] Zivkovic AM, German JB, Lebrilla CB, Mills DA. Human milk glycobiome 2011;6:e25880.

and its impact on the infant gastrointestinal microbiota. Proceedings of the

[39] Uehara Y, Kikuchi K, Nakamura T, Nakama H, Agematsu K, Kawakami Y, et al.

National Academy of Sciences of the United States of America 2011;108(Suppl

H2O2 produced by viridans group streptococci may contribute to inhibition

1):4653–8.

of methicillin-resistant Staphylococcus aureus colonization of oral cavities in

[15] Ward R, Ninonuevo M, Mills D, Lebrilla C, German J. In vitro fermentation

newborns. Clinical Infectious Diseases 2001;32:1408–13.

of breast milk oligosaccharides by Bifidobacterium infantis and Lactobacillus

[40] Diaz-Ropero MP, Martin R, Sierra S, Lara-Villoslada F, Rodríguez JM, Xaus J,

gasseri. Applied and Environment Microbiology 2006;72:4497–9.

et al. Two Lactobacillus strains, isolated from breast milk, differently mod-

[16] Sela DA, Chapman J, Adeuya A, Kim JH, Chen F, Whitehead TR, et al.

ulate the immune response. Journal of Applied Microbiology 2006;102:

The genome sequence of Bifidobacterium longum subsp. infantis reveals 337–43.

adaptations for milk utilization within the infant microbiome. Proceed-

[41] Olivares M, Díaz-Ropero MP, Gómez N, Lara-Villoslada F, Sierra S, Maldonado

ings of the National Academy of Sciences of the United States of America

JA, et al. The consumption of two new probiotic strains, Lactobacillus gasseri

2008;105:18964–9.

CECT5714 and Lactobacillus coryniformis CECT5711, boost the immune system

[17] Turroni F, Bottacini F, Foroni E, Mulder I, Kim JH, Zomer A, et al. Genome

of healthy adults. International Microbiology 2006;9:47–52.

analysis of Bifidobacterium bifidum PRL2010 reveals metabolic pathways for

[42] Olivares M, Díaz-Ropero MP, Sierra S, Lara-Villoslada F, Fonollá J, Navas M,

host-derived glycan foraging. Proceedings of the National Academy of Sci-

et al. Oral intake of Lactobacillus fermentum CECT5716 enhances the effects of

ences of the United States of America 2010;107:19514–9.

influenza vaccination. Nutrition 2007;23:254–60.

[18] Gavin A, Ostovar K. Microbiological characterization of human milk. Journal

[43] Pérez-Cano FJ, Dong H, Yaqoob P. In vitro immunomodulatory activity of

of Food Protection 1977;40:614–6.

Lactobacillus fermentum CECT5716 and Lactobacillus salivarius CECT5713:

[19] West PA, Hewitt JH, Murphy OM. The influence of methods of collection and

two probiotic strains isolated from human breast milk. Inmunobiology

storage on the bacteriology of human milk. Journal of Applied Bacteriology 2010;12:996–1004.

1979;46:269–77.

[44] Olivares M, Díaz-Ropero MP, Lara-Villoslada F, Rodriguez JM, Xaus J. Efec-

[20] Martín V, Manés-Lázaro˜ R, Rodríguez JM, Maldonado A. Streptococcus lactarius

tiveness of probiotics in allergy: child’s game or adult affair? Nutrafoods

sp. nov., isolated from breast milk of healthy women. International Journal of 2005;4:59–64.

Systematic and Evolutionary Microbiology 2011;61:1048–52.

[45] Kirjavainen PV, Apostolou E, Arvola T, Salminen SJ, Gibson GR, Isolauri E. Char-

[21] Matsumiya Y, Kato N, Watanabe K, Kato H. Molecular epidemiological study

acterizing the composition of intestinal microflora as a prospective treatment

of vertical transmission of vaginal Lactobacillus species from mothers to new-

target in infant allergic disease. FEMS Immunology and Medical Microbiology

born infants in Japanese, by arbitrarily primed polymerase chain reaction. 2001;32:1–7.

Journal of Infection and Chemotherapy 2002;8:43–9.

[46] Asakuma S, Hatakeyama E, Urashima T, Yoshida E, Katayama T, Yamamoto

[22] Albesharat R, Ehrmann MA, Korakli M, Yazaji S, Vogel RF. Phenotypic and

K, et al. Physiology of consumption of human milk oligosaccharides

genotypic analyses of lactic acid bacteria in local fermented food, breast milk

by infant gut-associated bifidobacteria. Journal of Biological Chemistry

and faeces of mothers and their babies. Systematic and Applied Microbiology 2011;286:34583–92.

2011;34:148–55.

[47] Maldonado J, Lara-Villoslada F, Sierra S, Sempere L, Gómez M, Rodriguez JM,

[23] Makino H, Kushiro A, Ishikawa E, Muylaert D, Kubota H, Sakai T, et al.

et al. Safety and tolerance of the human milk probiotic strain Lactobacil-

Transmission of intestinal Bifidobacterium longum subsp. longum strains from

lus salivarius CECT5713 in 6-month-old children. Nutrition 2010;26:1082–

mother to infant, determined by multilocus sequencing typing and ampli- 7.

fied fragment length polymorphism. Applied and Environment Microbiology

[48] Gil-Campos M, López MÁ, Rodriguez-Benítez MV, Romero J, Roncero I, Linares

2011;77:6788–93.

MD, et al. Lactobacillus fermentum CECT 5716 is safe and well tolerated in

[24] Martín V, Maldonado A, Moles L, Rodríguez-Banos˜ M, Del Campo R, Fernández

infants of 1-6 months of age: a randomized controlled trial. Pharmacological

L, et al. Sharing of bacterial strains between breast milk and infant feces.

Research 2012;65:231–8.

Journal of Human Lactation 2012;28:36–44.

[49] Ramsey DT, Kent JC, Owens RA, Hartmann PE. Ultrasound imaging of milk

[25] Hunt KM, Foster JA, Forney LJ, Schütte UME, Beck DL, Abdo Z, et al. Charac-

ejection in the breast of lactating women. Pediatrics 2004;113:361–7.

terization of the diversity and temporal stability of bacterial communities in

[50] Nasidze I, Li J, Quinque D, Tang K, Stoneking M. Global diversity in the human

human milk. PLoS ONE 2011;6:e21313.

salivary microbiome. Genome Research 2009;19:636–43.

[26] Sanz Y. Gut microbiota and probiotics in maternal and infant health. American

[51] Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Defining the normal

Journal of Clinical Nutrition 2011;94(Suppl 6):2000S–5S.

bacterial flora of the oral cavity. Journal of Clinical Microbiology 2005;43:

[27] Martín R, Heilig HG, Zoetendal EG, Jiménez E, Fernández L, Smidt H, et al. 5721–32.

Cultivation-independent assessment of the bacterial diversity of breast milk

[52] Yang F, Zeng X, Ning K, Liu KL, Lo CC, Wang W, et al. Saliva microbiomes

among healthy women. Research in Microbiology 2007;158:31–7.

distinguish caries-active from healthy human populations. ISME Journal

[28] Martín R, Heilig HG, Zoetendal EG, Smidt H, Rodríguez JM. Diversity of the 2012;6:1–10.

Lactobacillus group in breast milk and vagina of healthy women and potential

[53] Cephas KD, Kim J, Mathai RA, Barry KA, Dowd SE, Meline BS, et al. Compara-

role in the colonization of the infant gut. Journal of Applied Microbiology

tive analysis of salivary bacterial microbiome diversity in edentulous infants

2007;103:2638–44.

and their mothers or primary care givers using pyrosequencing. PLoS ONE

[29] Gueimonde M, Laitinen K, Salminen S, Isolauri E. Breast milk: a source 2011;6:e23503.

of bifidobacteria for infant gut development and maturation. Neonatology

[54] Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC. NISC Compara-

2007;92:64–6.

tive Sequencing Program, Bouffard GG, Blakesley RW, Murray PR, Green ED,

[30] Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, et al. Suc-

Turner ML, Segre JA. Topographical and temporal diversity of the human skin

cession of microbial consortia in the developing infant gut microbiome.

microbiome. Science 2009;324:1190–2.

Proceedings of the National Academy of Sciences of the United States of

[55] Gao Z, Tseng C, Pei Z, Blaser MJ. Molecular analysis of human forearm super-

America 2011;108(Suppl 1):4578–85.

ficial skin bacterial biota. Proceedings of the National Academy of Sciences of

[31] Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al.

the United States of America 2007;104:2927–32.

A core gut microbiome in obese and lean twins. Nature 2009;457:480–4.

[56] Martín R, Langa S, Reviriego C, Jiménez E, Marín ML, Olivares M, et al. The com-

[32] Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, McCulle SL, et al. Vagi-

mensal microflora of human milk: new perspectives for food bacteriotherapy

nal microbiome of reproductive-age women. Proceedings of the National

and probiotics. Trends in Food Science and Technology 2004;15:121–7.

L. Fernández et al. / Pharmacological Research 69 (2013) 1–10 9

[57] Rescigno M, Urbano M, Valzasina B, Francolín M, Rotta G, Bonasio R, et al. [86] Lawrence RA, Lawrence RM. Breastfeeding: a guide for the medical profession.

Dendritic cells express tight junction proteins and penetrate gut epithelial 6th ed. St. Louis: Elsevier Mosby; 2005.

monolayers to sample bacteria. Nature Immunology 2001;2:361–7. [87] Delgado S, Arroyo R, Jiménez E, Marín ML, del Campo R, Fernández L, et al.

[58] Vazquez-Torres A, Jones-Carson J, Baumler AJ, Falkow S, Valdivia R, Brown W, Staphylococcus epidermidis strains isolated from breast milk of women

et al. Extraintestinal dissemination of Salmonella by CD18-expressing phago- suffering infectious mastitis: potential virulence traits and resistance to

cytes. Nature 1999;401:804–8. antibiotics. BMC Microbiology 2009;9:82.

[59] Macpherson AJ, Uhr T. Induction of protective IgA by intestinal dendritic cells [88] Thorberg BM, Kuhn I, Aarestrup FM, Brandstrom B, Jonsson P, Danielsson-

carrying commensal bacteria. Science 2004;303:1662–5. Tham ML. Pheno- and genotyping of Staphylococcus epidermidis isolated from

[60] Roitt I. Essential immunology. Oxford: Blackwell Scientific Publications; 2001. bovine milk and human skin. Veterinary Microbiology 2006;115:163–72.

[61] Bertotto A, Gerli R, Castellucci G, Scalise F, Vaccaro R. Human milk lympho- [89] Zhang S, Maddox CW. Cytotoxic activity of coagulase-negative staphylococci

cytes bearing the gamma/delta T-cell receptor are mostly delta TCS1-positive in bovine mastitis. Infection and Immunity 2000;68:1102–8.

cells. Immunology 1991;74:360–1. [90] Thomsen AC, Mogensen SC, Love Jepsen F. Experimental mastitis in mice

[62] Langa S. Interactions between lactic acid bacteria, intestinal epithelial cells induced by coagulase-negative staphylococci isolated from cases of mas-

and immune cells. Development of in vitro models. PhD Thesis. Madrid: Com- titis in nursing women. Acta Obstetricia et Gynecologica Scandinavica

plutense University of Madrid; 2006. 1985;64:163–6.

[63] Langa S, Maldonado A, Delgado S, Martín R, Martín V, Jiménez E, et al. Charac- [91] Herold BC, Immergluck LC, Maranan MC, Lauderdale DS, Gaskin RE, Boyle-

terization of Lactobacillus salivarius CECT 5713, a strain isolated from human Vavra S, et al. Community-acquired methicillin-resistant Staphylococcus

milk: from genotype to phenotype. Applied Microbiology and Biotechnology aureus in children with no identified predisposed risk. Journal of the American

2012;94:1279–87. Medical Association 1998;279:593–8.

[64] Rescigno M, Citterio S, Thery C, Rittig M, Medaglini D, Pozzi G, et al. Bacteria- [92] Saiman L, O’Keefe M, Graham 3rd PL, Wu F, Saïd-Salim B, Kreiswirth B,

induced neo-biosynthesis, stabilization, and surface expression of functional et al. Hospital transmission of community-acquired methicillin-resistant

class I molecules in mouse dendritic cells. Proceedings of the National Staphylococcus aureus among postpartum women. Clinical Infectious Diseases

Academy of Sciences of the United States of America 1998;95:5229–34. 2003;37:1313–9.

[65] Perez PF, Doré J, Leclerc M, Levenez F, Benyacoub J, Serrant P, et al. Bacte- [93] Jones TF, Creech CB, Erwin P, Baird SG, Woron AM, Schaffner W. Family out-

rial imprinting of the neonatal immune system: lessons from maternal cells. breaks of invasive community-associated methicillin resistant Staphylococcus

Pediatrics 2007;119:e724–32. aureus infection. Clinical Infectious Diseases 2006;42:76–8.

[66] Jiménez E, Fernández L, Maldonado A, Martín R, Olivares M, Xaus J, et al. Oral [94] Reddy P, Qi C, Zembower T, Noskin GA, Bolon M. Postpartum mastitis and

administration of lactobacilli strains isolated from breast milk as an alter- community-acquired methicillin-resistant Staphylococcus aureus. Emerging

native for the treatment of infectious mastitis during lactation. Applied and Infectious Diseases 2007;13:298–301.

Environment Microbiology 2008;74:4650–5. [95] Naimi TS, LeDell KH, Como-Sabetti K, Borchardt SM, Boxrud DJ, Etienne J, et al.

[67] Arroyo R, Martín V, Maldonado A, Jiménez E, Fernández L, Rodríguez JM. Comparison of community- and health care-associated methicillin-resistant

Treatment of infectious mastitis during lactation: antibiotics versus oral Staphylococcus aureus infection. Journal of the American Medical Association

administration of lactobacilli isolated from breast milk. Clinical Infectious 2003;290:2976–84.

Diseases 2010;50:1551–8. [96] Kourtis AP, Butera S, Ibegbu C, Belec L, Duerr A. Breast milk and HIV-1:

[68] Beischer NA, Mackay EV, Colditz PB. Obstetrics and the newborn. Philadel- vector of transmission or vehicle of protection. Lancet Infectious Diseases

phia: W.B. Saunders Company; 1997. 2003;3:786–93.

[69] Vankerckhoven VV, Autgaerden TA, Huys G, Vancanneyt M, Swings J, [97] Kourtis AP, Jamieson DJ, de Vincenzi I, Taylor A, Thigpen MC, Dao H, et al.

Goossens H. Establishment of the PROSAFE collection of probiotic and human Prevention of human immunodeficiency virus-1 transmission to the infant

lactic acid bacteria. Microbial Ecology in Health and Disease 2004;16:131–6. through breastfeeding: new developments. American Journal of Obstetrics

[70] Ouwenhand AC, Saxelin M, Salminen S. Phenotypic differences between com- and Gynecology 2007;197(Suppl.):S113–22.

mercial Lactobacillus rhamnosus GG and L. rhamnosus strains recovered from [98] Lepage P, Munyakazi C, Hennart P. Breastfeeding and hospital mortality in

blood. Clinical Infectious Diseases 2004;39:1858–60. children in Rwanda. Lancet 1982;1:403.

[71] Jiménez E, Fernández L, Marín ML, Martín R, Odriozola JM, Nueno-Palop C, [99] WHO Collaborative Study Team on the Role of Breastfeeding on the Preven-

et al. Isolation of commensal bacteria from umbilical chord blood of healthy tion of Infant Mortality. Effect of breastfeeding on infant and child mortality

neonates born by caesarean section. Current Microbiology 2005;51:270–4. due to infectious diseases in less developed countries: a pooled analysis.

[72] Begier EM, Barrett NL, Mshar PA, Johnson DG, Hadler JL. Connecticut Bioter- Lancet 2000;355:451–5.

rorism Field Epidemiology Response Team. Gram-positive rod surveillance [100] WHO, UNICEF, UNFPA, UNAIDS. HIV and infant feeding: new evidence and

for early anthrax detection. Emerging Infectious Diseases 2005;11:1483–6. programmatic experience: report of a technical consultation. Geneva: World

[73] Dasanayake AP, Li Y, Wiener H, Ruby JD, Lee MJ. Salivary Actinomyces Health Organization; 2007.

naeslundii genospecies 2 and Lactobacillus casei levels predict pregnancy out- [101] Coutsoudis A, Pillay K, Spooner E, Kuhn L, Coovadia HM. For the South African

comes. Journal of Periodontology 2005;76:171–7. Vitamin A Study Group. Influence of infant-feeding patterns on early mother-

[74] Aso Y, Akaza H, Kotake T, Tsukamoto T, Imai K, Naito S. Preventive effect of a to-child transmission of HIV-1 in Durban, South Africa: a prospective cohort

Lactobacillus casei preparation on the recurrence of superficial bladder cancer study. Lancet 1999;354:471–6.

in a double-blind trial. European Urology 1995;27:104–9. [102] Kuhn L. Milk mysteries: why are women who exclusively breast-feed less

[75] Guarner F, Schaafsma GJ. Probiotics. International Journal of Food Microbiol- likely to transmit HIV during breast-feeding. Clinical Infectious Diseases

ogy 1998;39:237–8. 2010;50:770–2.

[76] Collins JK, Thornton G, O’Sullivan GO. Selection of probiotic strains for human [103] Lunney KM, Iliff P, Mutasa K, Ntozini R, Magder LS, Moulton LH, et al. Associ-

applications. International Dairy Journal 1998;8:487–90. ations between breast milk viral load, mastitis, exclusive breast-feeding, and

[77] Jiménez E, Langa S, Martín V, Arroyo R, Martín R, Fernández L, et al. Complete postnatal transmission of HIV. Clinical Infectious Diseases 2010;50:762–9.

genome sequence of Lactobacillus fermentum CECT 5716, a probiotic strain [104] Coovadia HM, Rollins NC, Bland RM, Little K, Coutsoudis A, Bennish ML, et al.

isolated from human milk. Journal of Bacteriology 2010;192:4800. Mother-to-child transmission of HIV-1 infection during exclusive breast-

[78] Jiménez E, Martín R, Maldonado A, Martín V. Gómez de Segura A, L. Fernández feeding in the first 6 months of life: an intervention cohort study. Lancet

L, Rodríguez JM. Complete genome sequence of Lactobacillus salivarius CECT 2007;369:1107–16.

5713, a probiotic strain isolated from human milk and infant feces. Journal of [105] Becquart P, Chomont N, Roques P, Ayouba A, Kazatchkine MD, Bélec L,

Bacteriology 2010;192:5266–7. et al. Compartmentalization of HIV-1 between breast milk and blood of HIV-

[79] Jiménez E, Villar-Tajadura MA, Marín M, Fontecha J, Requena T, Arroyo R, infected mothers. Virology 2002;300:109–17.

et al. Complete genome sequence of Bifidobacterium breve CECT 7263, a strain [106] Becquart P, Hocini H, Lévy M, Sépou A, Kazatchkine MD, Bélec L. Secre-

isolated from human milk. Journal of Bacteriology 2012;194:3762–3. tory anti-human immunodeficiency virus (HIV) antibodies in colostrum and

[80] Martín V, Maldonado-Barragán A, Jiménez E, Ruas-Madiedo P, Fernández L, breast milk are not a major determinant of the protection of early postnatal

Rodríguez JM. Complete genome sequence of Streptococcus salivarius PS4, a transmission of HIV. Journal of Infectious Diseases 2000;181:532–9.

strain isolated from human milk. Journal of Bacteriology 2012;194:4466–7. [107] Lyimo MA, Howell AL, Balandya E, Eszterhas SK, Connor RI. Innate factors

[81] Gdalevich M, Mimouni D, Mimouni M. Breast-feeding and the risk of bronchial in human breast milk inhibit cell-free HIV-1 but not cell-associated HIV-1

asthma in childhood: a systematic review with meta-analysis of prospective infection of CD4+ cells. Journal of Acquired Immune Deficiency Syndromes

studies. Journal of Pediatrics 2001;139:261–6. 2009;51:117–24.

[82] Gdalevich M, Mimouni D, David M, Mimouni M. Breast-feeding and the onset [108] Kuhn L, Trabattoni D, Kankasa C, Sinkala M, Lissoni F, Ghosh M, et al. HIV-

of atopic dermatitis in childhood: a systematic review with meta-analysis specific secretory IgA in breast milk of HIV-positive mothers is not associated

of prospective studies. Journal of the American Academy of Dermatology with protection against HIV transmission among breast-fed infants. Journal

2001;45:520–7. of Pediatrics 2006;149:611–6.

[83] Kalliomäki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. [109] Martín V, Maldonado A, Fernández L, Rodríguez JM, Connor RI. Inhibition of

Probiotics in primary prevention of atopic disease: a randomized placebo- human immunodeficiency virus type 1 by lactic acid bacteria from human

controlled trial. Lancet 2001;357:1076–9. breastmilk. Breastfeeding Medicine 2010;5:153–8.

[84] Foxman B, D’Arcy H, Gillespie B, Bobo JK, Schwartz K. Lactation mastitis: [110] Baba N, Samson S, Bourdet-Sicard R, Rubio M, Sarfati M. Commensal bacteria

occurrence and medical management among 946 breastfeeding women in trigger a full dendritic cell maturation program that promotes the expansion

the United States. American Journal of Epidemiology 2002;155:103–14. of non-Tr1 suppressor T cells. Journal of Leukocyte Biology 2008;84:468–76.

[85] World Health Organization. Mastitis: causes and management. Geneva: [111] Konstantinov SR, Smidt H, de Vos WM, Bruijns SC, Singh SK, Valence F,

WHO; 2000. et al. S layer protein A of Lactobacillus acidophilus NCFM regulates immature

10 L. Fernández et al. / Pharmacological Research 69 (2013) 1–10

dendritic cell and T cell functions. Proceedings of the National Academy of [117] Gori A, Tincati C, Rizzardini G, Torti C, Quirino T, Haarman M, et al. Early

Sciences of the United States of America 2008;105:19474–9. impairment of gut function and gut flora supporting a role for alteration

[112] Mohamadzadeh M, Olson S, Kalina WV, Ruthel G, Demmin GL, Warfield KL, of gastrointestinal mucosa in human immunodeficiency virus pathogenesis.

et al. Lactobacilli activate human dendritic cells that skew T cells toward T Journal of Clinical Microbiology 2008;46:757–8.

helper 1 polarization. Proceedings of the National Academy of Sciences of the [118] Coutsoudis A. Influence of infant feeding patterns on early mother-to-child

United States of America 2005;102:2880–5. transmission of HIV-1 in Durban, South Africa. Annals of the New York

[113] Lo P. HIV hijacks dendritic cells. Nature Medicine 2003;9:650. Academy of Sciences 2000;918:136–44.

[114] Melki MT, Saïdi H, Dufour A, Olivo-Marin JC, Gougeon ML. Escape of HIV- [119] Smith MM, Kuhn L. Exclusive breast-feeding: does it have the poten-

1-infected dendritic cells from TRAIL-mediated NK cell cytotoxicity during tial to reduce breast-feeding transmission of HIV-1. Nutrition Reviews

NK-DC cross-talk-a pivotal role of HMGB1. PLoS Pathogens 2010;4:e1000862. 2000;58:333–40.

[115] Chang R, Pavlova S, Caffrey M, Spear G, Tanzer J, Thompson A, et al. Blocking [120] Sinkiewicz G, Ljunggren L. Occurrence of Lactobacillus reuteri in

milk-borne HIV transmission by virus-capturing lactobacilli. In: The mouth human breast milk. Microbial Ecology in Health and Disease 2008;20:

and AIDS: the global challenge 6th world workshop on oral health and disease. 122–6.

2009. [121] Collado MC, Delgado S, Maldonado A, Rodríguez JM. Assessment of the bacte-

[116] Catassi C, Bonucci A, Coppa GV, Carlucci A, Giorgi PL. Intestinal permeabil- rial diversity of breast milk of healthy women by quantitative real time PCR.

ity changes during the first month: effect of natural versus artificial feeding. Letters in Applied Microbiology 2009;48:523–8.

Journal of Pediatric Gastroenterology and Nutrition 1995;21:383–6.