Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Journal of Functional Foods 37 (2017) 74–89

Contents lists available at ScienceDirect

Journal of Functional Foods


journal homepage: www.elsevier.com/locate/jff

Streptococcus thermophilus: From yogurt starter to a new promising


probiotic candidate?
Ophélie Uriot a,b, Sylvain Denis a, Maira Junjua b, Yvonne Roussel b, Annie Dary-Mourot b,⇑,1,
Stéphanie Blanquet-Diot a,1
a
UMR 454 UCA-INRA MEDIS Microbiota, Digestive Environment and Health, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
b
URAFPA, Unité de Recherche Animal et Fonctionnalités des Produits Animaux, Equipe Protéolyse et Biofonctionnalité des Protéines et des Peptides, Université de Lorraine,
54506 Vandoeuvre-les-Nancy, France

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

Article history: Probiotics are defined as live microorganisms that when administered in adequate amount confer a
Received 10 March 2017 health benefit to the host. To be considered as a probiotic, a bacterial strain must not only be safe but
Received in revised form 7 July 2017 should also survive in the human gastrointestinal tract and exert health benefits on its host.
Accepted 18 July 2017
Streptococcus thermophilus is a Gram positive bacterium widely used in dairy fermentations for the
production of yogurt and cheese. In contrast with other lactic acid bacteria, the probiotic status of
S. thermophilus remains still questioned. This review gives an update of the human trials, in vivo assays
Keywords:
in animal models and in vitro experiments, which have assessed the resistance of S. thermophilus to gas-
S. thermophilus
Probiotic
trointestinal stresses and have investigated its positive health effects. The underlying mechanisms of
Gastrointestinal survival action are also described and the probiotic status of the bacterium is debated with respect to the available
Health effects literature.
Ó 2017 Published by Elsevier Ltd.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
2. S. thermophilus: a key LAB in dairy production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3. Survival of S. thermophilus in the digestive environment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.1. Human trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.2. In vivo studies in animal models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.3. In vitro experiments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
4. Acid and bile resistance mechanisms in S. thermophilus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5. Beneficial health effects of S. thermophilus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.1. Alleviation of lactose intolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.2. Prevention of chronic gastritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.3. Prevention of diarrhea . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
6. Possible mechanisms of action of S. thermophilus on the host health. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
6.1. Antimicrobial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
6.2. Antioxidant activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
6.3. Immunomodulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
6.4. Effect on intestinal barrier function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
6.5. Interactions with intestinal microbiota . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86

⇑ Corresponding author at: URAFPA, Université de Lorraine, Faculté des Sciences et Techniques, Campus Aiguillettes, 54506 Vandoeuvre-les-Nancy, France.
E-mail address: annie.dary@univ-lorraine.fr (A. Dary-Mourot).
1
Co-senior authors.

http://dx.doi.org/10.1016/j.jff.2017.07.038
1756-4646/Ó 2017 Published by Elsevier Ltd.
O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89 75

1. Introduction health to patients suffering from a form of malnutrition caused by


the consumption of highly refined food (Kollath, 1953). One year
The original hypothesis of the positive role played by certain later, Ferdinand Vergin proposed that probiotics were the opposite
bacteria was first introduced by Élie Metchnikoff, who wrote in of antibiotics (Vergin, 1954). In 1965, the work of Lilly and Still-
1907 that ‘‘A reader who has little knowledge about such matters well (Lilly & Stillwell, 1965) gave a new dimension to the defini-
may be surprised by my recommendation to absorb large quantities tion of probiotics. Observations that some secretions of
of microbes, as a general belief is that microbes are harmful. This Colpidium campylum could increase the growth of Tetrahymena
belief is erroneous. There are many useful microbes, amongst which pyriformis prompted them to define probiotic as ‘‘the secretions
the lactic bacilli have honorable place.” and ‘‘The dependence of the of one microorganism which stimulate the growth of another
intestinal microbes on the food makes it possible to adopt measures microorganism”. Similarly, in 1971, the term probiotics referred
to modify the flora in our bodies and to replace the harmful microbes to tissue extracts capable of stimulating microbial growth
by useful microbes.” (Metchnikoff, 1907). From Metchnikoff till (Sperti, 1971). In 1974, the word probiotic was used for the first
today, the probiotic concept has drastically evolved and different time to describe a microbial feed/food supplement and defined
definitions of probiotics have been proposed (Table 1). For exam- as ‘‘organisms and substances contributing to the intestinal micro-
ple, in 1953, the word ‘‘probiotika” was used by Werner Kollath to bial balance” (Parker, 1974). In 1992, Fuller removed the word
designate organic and inorganic supplements necessary to restore ‘‘substances” from the probiotic’s definition and emphasized on
their viability defining probiotics as ‘‘live microbial feed supple-
ments which beneficially affect the host animal by improving
Table 1
Evolutionary stages of probiotic definitions. Adapted from Vasiljevic and Shah (2008) microbial balance” (Fuller, 1992). Because this definition was not
wide enough to cover the entire indigenous microflora, another
Year Description Sources
definition was proposed by Havenaar and Huis in‘t Veld in 1992
1907 First hypothesis of a positive role for Metchnikoff (1907) (Havenaar & Huis in’t Veld, 1992). In their view, ‘‘probiotics are
certain bacteria
the viable mono- or mixed microbial cultures which when applied
1953 First use of the word ‘‘probiotika” which Kollath (1953)
appoints organic and inorganic to animal or man have beneficial effects on their host by improv-
supplements necessary to restore health ing the properties of indigenous microflora”. In 1996, the cultured
1954 Probiotics are defined as opposite of Vergin (1954) dairy products were also added to the probiotic concept and
antibiotics Schaafsma defined them as ‘‘living microorganisms which when
1955 Probiotics can be effective against the Kolb (1955)
ingested in certain numbers exert health benefits beyond inherent
deleterious effects of antibiotics
1965 Substances secreted by one microorganism Lilly and Stillwell basic nutrition” (Schaafsma, 1996). This definition was confirmed
which stimulate the growth of another one (1965) in 2002 by the Food and Agriculture Organization (FAO) of the
1971 Tissue extracts able to stimulate microbial Sperti (1971) United Nations and World Health Organization working group
growth
(FAO/WHO, 2002) stating that probiotics are ‘‘live microorganisms
1973 Compounds that build resistance to Fujii and Cook (1973)
infections in the host but do not inhibit the
which when administered in adequate amounts, confer a health
growth of microorganisms in vitro benefit on the host”, and reconfirmed by the FAO in 2006. In
1974 Organisms and substances contributing to Parker (1974) 2014, this definition was revised with minor grammatical correc-
microbial balance in the intestine tion by an expert panel of the International Scientific Association
1992 Live microbial feed supplements which Fuller (1992)
for Probiotics and Prebiotics as, ‘‘live microorganisms that, when
beneficially affect the host animal by
improving microbial balance administrated in adequate amounts, confer a health benefit on
1992 Viable mono- or mixed culture of live Havenaar and Huis in’t the host” (Hill et al., 2014). In 2015, a framework was also pro-
microorganisms which, when applied to Veld (1992) posed for the characterization of novel probiotic bacteria and the
animals or mans, have a beneficial effect on
assessment of their safety and efficacy in accordance with Euro-
the host by improving the properties of the
indigenous microflora
pean health policy (Miquel et al., 2015).
1996 Live microbial culture or cultured dairy Salminen (1996) Numerous health benefits resulting from the ingestion of probi-
products which beneficially influence the otics have been observed in animal models and are expected to
health and nutrition of the host occur in humans as well, while others have been specifically
1996 Living microorganisms which, upon Schaafsma (1996)
proved through human trials (Table 2). These health benefits are
ingestion in certain numbers, exert health
benefits beyond inherent basic nutrition mainly associated with the prevention and treatment of diarrhea
1999 Microbial cell preparations or components Salminen, Ouwehand, symptoms, the prevention of irritable bowel diseases, colitis and
of microbial cells that have a beneficial Benno, and Lee (1999) necrotizing enterocolitis and the relief in lactose intolerance (Di
effect on the health and well-being of the
Cerbo, Palmieri, Aponte, Morales-Medina, & Iannitti, 2016; Hill
host
2001 A preparation or a product containing Schrezenmeir and de
et al., 2014; Nagpal et al., 2012; Vasiljevic & Shah, 2008). In addi-
viable, defined microorganisms in Vrese (2001) tion, certain probiotic bacteria have been recommended for the
sufficient numbers, which alter the treatment of extra-intestinal pathologies such as atopic dermatitis,
microflora (by implantation or hypercholesterolemia or allergies (Boyle & Tang, 2006; Di Cerbo
colonization) in a compartment of the host
et al., 2016; Vasiljevic & Shah, 2008). Overall, these health effects
and in this way exert beneficial health
effects on this host are highly strain-dependent and there is not a single probiotic
2002 Live microorganisms which when FAO/WHO (2002) strain which can confer all the benefits previously reported
administered in adequate amounts, confer (Senok, Ismaeel, & Botta, 2005; Shah, 2007). Even if certain mech-
a health benefit on the host anism action of probiotics begin to be well described in the litera-
2014 Grammatical adjustment of the FAO/WHO Hill et al. (2014
definition: live microorganisms that, when
ture, such as the anti-pathogenic activities, which include direct
administered in adequate amounts, confer antagonism, immunomodulation and competitive exclusion
a health benefit on the host (Preidis et al., 2011), they are far from being fully elucidated. How-
2015 List of criteria that should be assessed to Miquel et al. (2015) ever, it is yet known that after their consumption, probiotics may
obtain marketing authorization and health
exert their beneficial effects by influencing the intestinal luminal
claim for probiotic bacteria in accordance
with European health policy environment, the epithelial and mucosal barrier function as well
as the immune system (Nagpal et al., 2012).
76 O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89

Table 2
Main bacterial and yeast probiotics and their health effects, as observed in animal models or during human trials.

Effects on health Strains Human Animal References


trials models
Prevention of diarrhea B. bifidum (LMG-P17500) X – Beniwal et al. (2003), Bowen et al. (2007), Canani et al. (2007), Di Cerbo et al.
B. breve (K-110) X X (2016), Korpela et al. (2016), O’Callaghan and van Sinderen (2016), Saavedra et al.
B. infantis (CECT7210) X X (1994), Saavedra et al. (2004), Shah (2007), and Trabelsi, Ktari, Ben Slima, Hamden,
B. lactis (B94; Bb12) X – and Ben Salah (2016)
B. longum (SBT-2828) X X
Lb. acidophilus (CERELA; X X
DSM21007; GP1B; La-5; LMG-
P17549)
Lb. bulgaricus (LMG-P17550) X X
Lb. casei (CERELA; DN-114001; X X
Shirota)
Lb. fermentum (353; KLD) X –
Lb. plantarum (2017405; TN8) X X
Lb. reuteri (DSM-12246; DSM- X –
17938)
Lb. rhamnosus (35; E/N; GG; X X
Oxy; Pen)
S. boulardii (It) X –
S. thermophilus (LMG-P17503) X X
Prevention of B. breve (M-16 V) X – Guandalini et al. (2010), O’Callaghan and van Sinderen (2016), and Shah (2007)
inflammatory bowel B. longum X –
diseases B. infantis X –
Lb. acidophilus X –
Lb. plantarum X X
Lb. casei X –
Lb. bulgaricus X –
S. thermophilus X –
Prevention of gastric B. bifidum X – Di Cerbo et al. (2016), Hauser, Salkic, Vukelic, JajacKnez, and Stimac (2015), Hurduc,
diseases and H. pylori B. breve (Bb99) X – Plesca, Dragomir, Sajin, and Vandenplas (2009), Shah (2007), and Wang and Huang
infections B. lactis (Bb12) X – (2014)
B. longum (HY8001) X –
Lb. acidophilus (HY2177; La5; X –
LB; NAS)
Lb. casei (DG; DN-114001; X –
HY2743, Shirota)
Lb. gasseri (OLL2716) X –
Lb. johnsonii (La1; No1088) X X
Lb. paracasei (F19) X –
Lb. reuteri (ATCC55730; X –
ATCCPTA6475; DSM17938)
Lb. rhamnosus (GG; LC705) X –
P. freundenreichii subsp. X –
shermanii (JS)
S. boulardii X –
S. thermophilus (B-1) X –
Oral health effect Lb. brevis (CD2) X – Campus et al. (2014), Hasslöf, West, Videhult, Brandelius, and Stecksén-Blicks
Lb. paracasei (F19; SD1) X – (2013), Näse et al. (2001), Stensson et al. (2014), Teanpaisan and Piwat (2014)
Lb. reuteri (ATCC55730) X –
Lb. rhamnosus (GG) X –
Alleviation of lactose Lb. bulgaricus X – EFSA (2010)
intolerance S. thermophilus X –
Prevention of respiratory B. lactis (Bi-07) X – Di Cerbo et al. (2016); Guillemard, Tondu, Lacoin, and Schrezenmeir (2010), Leyer,
tract infections Lb. acidophilus (NCFM) X – Li, Mubasher, Reifer, and Ouwehand (2009), and Makino et al. (2010)
Lb. bulgaricus (OLL1073 R-1) X –
Lb. casei (DN-114001) X –
Lb. pentosus (b240) X –
Prevention of urogenital Lb. acidophilus (LA02) X – Di Cerbo et al. (2016)
infections Lb. brevis (CD2) X –
Lb. fermentum (LF10; LF15; X –
LN99; RC-14)
Lb. gasseri (LN40; Lba EB01- X –
DSM 14869)
Lb. plantarum (FV9; LP01; X –
P17630)
Lb. reuteri (CRL1324) X –
Lb. rhamnosus (GR-1; Lbp X –
PB01-DSM 14870; LN113)
Lb. salicinius (FV2) X –
P. acidilactici (LN23) X –
O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89 77

Table 2 (continued)

Effects on health Strains Human Animal References


trials models
Prevention of allergy and B. adolescentis X – Di Cerbo et al. (2016), Giovannini et al. (2007), Koyama et al. (2010), Shah (2007),
dermatitis B. bifidum X – West, Hammarström, and Hernell (2009)
B. breve (BR03) X –
Lb. acidophilus (L-92) X –
Lb. casei (DN-114001; Shirota) X –
Lb. gasseri (SBT0270;SBT0274) – X
Lb. paracasei (F19; LP-33) X –
Lb. reuteri (CRL1098) – X
Lb. rhamnosus (GG; GR-1) X –
Lb. salivarius (LS01) X –
Antimutagenic and B. animalis (DN-173010) – X Di Cerbo et al. (2016), O’Callaghan and van Sinderen (2016), and Shah (2007)
anticarcigenic B. lactis (Bb12; LAFTI-B94) X X
properties B. infantis (ATCC15697) – X
B. longum (25; 913; BB536) – X
E. faecalis X –
Lb. acidophilus (145; LAFTI- X X
L10; NCFM)
Lb. brevis (CD2) X –
Lb. bulgaricus (2 9 1) – X
Lb. casei (CRL431) – X
Lb. rhamnosus (GG; LC705) X X
P. freundenreichii subsp. X –
shermanii
S. thermophilus (DN-001158; – X
F4; V3)
Antioxidant activities B. bifidum (ATCC29521) – X Bouhafs, Moudilou, Exbrayat, Lahouel, and Idoui (2015), Di Cerbo et al. (2016), Ito
B. lactis (DSMZ23032) – X et al. (2015), Ito et al. (2008), Ito et al. (2003), Terahara et al. (2001), Tian et al.
B. longum (51 A) – X (2015), Vieira et al. (2015), and Wang, Xu, Xu, Zeng, and Wei (2016)
Lb. acidophilus (DSMZ23033) – X
Lb. brevis (DSMZ23034) – X
Lb. bulgaricus (2038) – X
Lb. plantarum (BJ0021) – X
Lb. rhamnosus (CCFM1107) – X
S. thermophilus (YIT2001) – X
Modulation of the B. breve (YIT4064) – X Ogita et al. (2015), Palma et al. (2015), and Shah (2007)
immune system Lb. casei (Shirota) – X
Lb. rhamnosus (OLL2838) – X
S. boulardii (LV01) – X
S. cerevisiae (CNCMI-3856; – X
LV02; UFMG905)
Hypocholesterolemic B. lactis X – Di Cerbo et al. (2016)
effect Lb. acidophilus (L1) X –
Lb. plantarum (CECT7527; X –
CECT7528; CECT7529)
Lb. reuteri (NCIMB 30242) X –

It is widely admitted that probiotics must survive through the Bifidobacterium (e.g., Bifidobacterium (B.) lactis, B. bifidum and B.
human gastrointestinal tract (GI) to exert their beneficial health breve) or Enterococci (e.g., Enterococcus (E.) faecalis) and some spe-
effects. Therefore, they have to resist to GI stresses, such as low cies of fungi (e.g., Saccharomyces (S.) boulardii and S. cerevisiae).
pH, bile salts and pancreatic secretions. Besides, the dose required Among LAB, Streptococcus (S.) thermophilus or Lb. delbrueckii subsp.
for a probiotic effect is still debated, due to fluctuations depending bulgaricus, which are traditionally used in yogurt production, have
on both the strain and target. Nevertheless, the minimal concentra- not been considered as probiotics during a long time by most sci-
tion of cells needed to obtain a clinical effect was quoted to be entists as they were not found to survive in the GI tract (Senok
106 CFU/mL in the small bowel and 108 CFU/mL in the colon et al., 2005). However, in 2010, yogurt which is the product of milk
(Minelli & Benini, 2008). Probiotics can be commercialized as fermentation by these two bacteria has obtained the European
freeze dried or encapsulated microorganisms in form of tablet, cap- Food Safety Agency (EFSA) scientific substantiation of health claim
sule or powder or they can be integrated into food products for lactose digestion improvement. The target population is indi-
(Foligné, Daniel, & Pot, 2013). Hitherto, fermented food and espe- viduals with lactose maldigestion and the yogurt should contain
cially dairy products have been the main vectors of probiotic at least 108 CFU live starter microorganisms per gram of fermented
administration to humans (Nagpal et al., 2012). The mode of probi- product (EFSA, 2010). Even if yogurt properties are recognized by
otic administration must be carefully considered as it can highly EFSA, the probiotic status of S. thermophilus itself is still
affect the persistence of the probiotic in the human digestive tract questioned.
(Blanquet-Diot et al., 2012; Pitino et al., 2012; Possemiers, In this context, the aim of the present review is to give an
Marzorati, Verstraete, & Van de Wiele, 2010). update on the probiotic status of S. thermophilus by summarizing
Many of probiotics currently used come from the human intes- the current published information related to its ability to survive
tine. Among them, are lactic acid bacteria (LAB) such as Lactobacil- in the digestive environment and its beneficial effects on host
lus (e.g., Lactobacillus (Lb.) acidophilus, Lb. rhamnosus and Lb. casei), health.
78 O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89

2. S. thermophilus: a key LAB in dairy production since they must meet very strict criteria. The survival of S. ther-
mophilus during transit through the human digestive environment
S. thermophilus is a Gram-positive bacterium showing ovoid remained controversial for a long period. Several studies suggested
cells occurring in pairs or in short chains. It is a thermophilic bac- that S. thermophilus did not survive the passage of the human GI
terium with an optimal growth temperature of 42 °C and an aero- tract (Ballesta, Velasco, Borobio, Argüelles, & Perea, 2008; del
tolerant anaerobe organism. S. thermophilus belongs to the salivar- Campo et al., 2005; Pedrosa et al., 1995) while other ones showed
ius group which also includes S. salivarius and S. vestibularis the opposite (Elli et al., 2006; García-Hernández, Moreno, Chuan, &
(Facklam, 2002; Gao, Zhi, Li, Klenk, & Li, 2014). The genome of S. Hernández, 2012; Mater et al., 2005; Pochart, Dewit, Desjeux, &
thermophilus is 1.8 Mb (Bai et al., 2016; Bolotin et al., 2004; Bourlioux, 1989; Venturi et al., 1999). In the studies from
Makarova et al., 2006; Sun et al., 2011), making it among the small- Pedrosa et al. (1995), del Campo et al. (2005) and Ballesta et al.
est genomes by comparison to other LAB and other Streptococcus (2008), fecal samples were collected from volunteers after con-
strains. Genetic studies indicate that S. thermophilus individualized sumption of yogurt. Detection of S. thermophilus was done by agar
from other pathogenic Streptococcus species about 7000 years ago plating or by specific polymerase chain reaction (PCR), but no cell
(Delorme, Bartholini, Bolotine, Ehrlich, & Renault, 2010; could be detected in the feces of volunteers. On the contrary,
Rasmussen et al., 2008). Since the S. thermophilus species diverged Pochart et al. (1989) established that S. thermophilus S85 had the
from their pathogenic relatives, it has lost most of the genes capacity to survive the passage through the upper GI tract, with
responsible for virulence and have acquired several genes useful at least 109 of the total 1011 ingested cells that survived in the
for living in milk (Hols et al., 2005). Analysis of different S. ther- stomach and reach the small intestine. Other studies detected
mophilus genomes revealed genes acquired through horizontal viable S. thermophilus in the feces of volunteers at concentrations
transfer which encode various functions, such as bacteriocin ranging between 3.5 and 5.6 log10 CFU/g of feces after ingestion
biosynthesis, efflux/uptake pumps, cell-envelope protease and pro- of 10–11 log10 CFU (Elli et al., 2006; García-Hernández et al.,
teins involved in peptide uptake and exopolysaccharide biosynthe- 2012; Mater et al., 2005; Venturi et al., 1999). During consumption,
sis (Bolotin et al., 2004; Delorme et al., 2010; Jameh et al., 2016; the concentration of viable S. thermophilus in the feces rapidly
Liu, Siezen, & Nauta, 2009; Rasmussen et al., 2008). increased to reach a plateau. Yet, one or two weeks after suspen-
S. thermophilus is the only Streptococcus species used in food sion of yogurt consumption, S. thermophilus counts dropped below
industry. Because it has been consumed by humans for centuries the detection limit (García-Hernández et al., 2012; Venturi et al.,
without giving any disease, it is also the only Streptococcus species 1999). Similar results were obtained using a PCR-detection
to be recognized as a Generally Recognized As Safe bacterium by method, with S. thermophilus disappearing completely from feces
the Food and Drug Administration (FDA). S. thermophilus is one of 3–9 days after stopping consumption (Brigidi, Swennen, Vitali,
the basic starter bacteria of yogurt and is the second most impor- Rossi, & Matteuzzi, 2003). At least two different elements may
tant species of industrial LAB after Lactococcus lactis. Besides the explain the contradictory results between the detection and the
traditional use of S. thermophilus in preparation of yogurt, it is used non-detection of viable S. thermophilus in feces presented above:
in production of several cheese varieties such as Emmental, the type of agar medium used to detect S. thermophilus living cells
Camembert, Brie, Mozzarella and Parmesan (Fox, McSweeney, from feces, and the strains used in these experiments. Indeed, Elli
Cogan, & Guinee, 2004; Hols et al., 2005). In yogurt production, et al. (2006) have shown that the detection of S. thermophilus in
the main role of S. thermophilus is a rapid acidification related to feces was strongly dependent on the medium used, and it was
lactic acid production, but also the production of secondary fer- observed in vitro (see below) that the survival of S. thermophilus
mentation products such as formate (Perez, de Antoni, & Añon, in the digestive tract strongly varied from one strain to another
1991), acetaldehyde or diacetyle which contribute to the aromatic (Junjua et al., 2016; Uriot et al., 2016).
and textural properties of the fermented products. In yogurt starter Even if these studies showed that S. thermophilus can survive
cultures, interactions between S. thermophilus and Lb. bulgaricus the passage through the human GI tract and be found alive in
are described as proto-cooperation (Hols et al., 2005), which is a the feces of people after consumption, it quickly disappears after
symbiotic relationship between the two species. S. thermophilus cessation of ingestion, in comparison with other LAB that can be
produces CO2 and formic acid which stimulate the growth of Lb. found longer in the feces (Venturi et al., 1999). In addition, as most
bulgaricus while Lb. bulgaricus hydrolyses milk proteins releasing of studies in humans were performed from fecal samples, very few
peptides and amino acids that improve S. thermophilus growth data are available on the survival of S. thermophilus throughout the
(Sieuwerts, de Bok, Hugenholtz, & van Hylckama Vlieg, 2008). In different compartments of the human digestive tract. Besides, none
cheese industry, S. thermophilus is mainly used in the refining of of these studies has investigated the survival of the bacterium
hard cheeses through the production of aromatic compounds from when ingested alone (only within a yogurt or a mix of probiotic
amino acids due to its glutamate dehydrogenase activity (Helinck, bacteria) and it is most of time impossible to calculate a specific
Le Bars, Moreau, & Yvon, 2004). survival rate for S. thermophilus based on published data in
humans.

3. Survival of S. thermophilus in the digestive environment 3.2. In vivo studies in animal models

For a probiotic strain, survival in the human GI tract is often Animal experimentation provides a good alternative to human
considered as a key factor to ensure the assigned health effect. clinical studies and can be used to predict what would happen in
Studies on the survival of S. thermophilus in the digestive environ- human digestive environment and apprehend what mechanisms
ment were performed either in humans, in animals or in in vitro are involved. However, animal digestive physiology remains differ-
models (Table 3). ent from that of human and the results obtained in animals cannot
be directly transposed to humans. To determine the capacity of S.
3.1. Human trials thermophilus to survive under digestive conditions, different ani-
mal models were used: minipigs (Lick, Drescher, & Heller, 2001),
To investigate the survival of probiotics in human digestive rabbits (Dilmi-Bouras & Sadoun, 2002), mice (Drouault, Anba, &
environment, clinical trials are obviously the golden standard. Corthier, 2002; Iyer, Tomar, Kapila, Mani, & Singh, 2010; Mater
However, these studies remain costly and heavy to implement et al., 2006) and rats (Ben-Yahia, Mayeur, Rul, & Thomas, 2012;
Table 3
Overview of S. thermophilus survival studies in the digestive environment: human trials, in vivo assays in animal models and in vitro experiments.

Studies Materiels and methods Results


Human Subjects Feeding strategies Evaluation of survival Key results
Pochart et al. (1989) 10 healthy adults Single intake of 430 g of Viable count by plating from 1.6  103 bacteria/g detected at 90 min in all the volunteers. 109 cells of the total 1011
yogurt (strain S85) duodenal samples ingested cells survived the stomach and reached the small intestine
Pedrosa et al. (1995) 33 elderly peoples 240 g of yogurt twice a day, Viable count by plating from No viable S. thermophilus detected in the gastric, duodenal and fecal samples
during 12 days gastric, duodenal and fecal
samples
Venturi et al. (1999) 20 UCr patients 3 g of VSL-3 twice a day, Viable count by plating from 7.88 log10 CFU/g of feces reached at days 20 and then remaining stable throughout the
during 1 year fecal samples study; disappeared 15 days after treatment suspension
Brigidi et al. (2003) 10 healthy adults 250 g of yogurt/day or 6 g of Direct quantitative PCR Healthy patients: reached 4  105 cells/g of feces (3 days, yogurt) or 5  106 cells/g of
10 IBD-patients VSL-3/day, during 10 days detection in fecal samples feces (13 days, VSL-3); disappeared 6–9 days after treatment suspension. IBD-patients:
or 2 months reached 107 cells/g of feces after 2 months (VSL-3)
del Campo et al. (2005) 114 healthy adults 375 g/day of pasteurized or PCR detection in fecal samples No S. thermophilus detected in the fecal samples
fresh yogurt during 2 weeks
Mater et al. (2005) 13 healthy adults 125 mL of yogurt twice a Viable count by plating from 6.3  104 CFU/g of feces detected in 82% of samples
day, during 12 days (strain fecal samples
S85)

O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89


Elli et al. (2006) 20 healthy adults 125 g of yogurt twice a day, Viable count by plating, PCR 5.6 log10 CFU/g of feces detected on day 7 in only one subject
during 1 week detection in fecal samples
Ballesta et al. (2008) Healthy adults Pasteurized or fresh yogurt, Viable count by plating, DNA No viable S. thermophilus detected in the fecal samples
during 75 days detection in fecal samples
García-Hernánez et al. 30 healthy adults 250 g of yogurt/day, during Viable count by DVC-FISH from Between 3.11 and 3.72 log10 CFU/g detected in week 2, between 3.46 and 4.48 log10 CFU/g
(2012) 4 weeks (strain CECT801) fecal samples in week 3 and between 3.76 and 4.66 log10 CFU/g in week 4; disappeared 4 weeks after
treatment suspension
Animal Models Feeding strategies Evaluation of survival Key results
Lick et al. (2001) 6 fistulated Göttingen minipigs Single intake of 600 g Viable count by plating and PCR Between 106 and 107 CFU/g of digesta detected between 3 and 6 h; disappeared rapidly
yogurt (strain 55n) detection from ilea digesta after 8 h
Dilmi-Bouras and 12 male rabbits 10 mL of yogurt twice a day, Viable count by plating from 5.2  108 CFU/mL of gastric samples and 6  106 CFU/mL of duodenal samples reached
Sadoun (2002) during 4 days gastric, duodenal and fecal after 1 h and 2 h, resp., and remaining stable throughout the study; 9.0  107 CFU/mL
samples reached after 72 h in fecal samples; disappeared 96 h after treatment suspension
Drouault et al., 2002 6 GF-mice Single intake of 0.5 mL milk Viable count by plating from Did not multiply and did not settle in GI tract of mice, only transited with the meal
cultures (strain FBI3) fecal samples
Mater et al. (2006) GF- and HMA-mice Single intake of 0.5 mL milk Viable count by plating from 107–108 CFU/g reached after 2.5 h then disappeared after 10 h in the small intestine and
cultures (strain FBI3) small intestine and colon 106–107 CFU/g reached after 2.5 h in the colon of the GF-mice; 107–108 CFU/g reached
samples after 1 h then disappeared after 6 h in the small intestine and reached 107 CFU/g after
2.5 h in the colon of the HMA-mice
Iyer et al. (2010) 32 male albino mice 30 g of fermented milk/day Viable count by plating and PCR No viable cell detected in the small intestine; 5 log10 CFU/g detected in the large intestine
during 20 days (strains detection in small and large and 8 log10 CFU/g in the feces; disappeared gradually after the end of the feeding period
RD102 and RD104) intestine and fecal samples
Thomas et al. (2011) 19 GF-rats Single intake of 1 mL milk Viable count by plating from Colonized the ileum and colon; 109–1010 CFU/g of feces reached in week 1 with lactose
cultures (strains LMD-9 and fecal samples and 108–109 CFU/g of feces in week 3 without lactose
LMG18311)
Ben-Yahia et al. (2012) 23 male GF-rats Single intake of 1 mL of Viable count by plating from Colonized the GI tract without lactose: progressively to 2  108 CFU/g of feces; with
milk cultures (strain LMD- fecal samples lactose: enhancement to 7.1  109 CFU/g of feces
9)
In vitro assays Models Tested strains Evaluation of survival Key results
Marteau et al. (1997) TIM model: dynamic stomach and ST20 Viable count by plating Survival decreased rapidly and dropped below 1% after 70 min in the gastric
small intestinal model; kinetics of compartment; 12% survival at 70 min in the duodenal compartment and <5% survival at
gastric and intestinal pH; 6 h 120 min in the ileal compartment, then the count remained stable
digestion at 37 °C
Vinderola and Simulated gastric juice: pH 2 or 3. 8 strains Viable count by plating Loss of 4–5.3 log10 CFU/mL for 6 strains and >6 log10 CFU/mL for 2 strains at pH 3; loss of
Reinheimer (2003) Bile salts: 0.3, 0.5 or 1%; incubation >6 log10 CFU/mL for the 8 strains at pH 2 (3 h); inhibition of growth at 0.5% and 1% of bile
37 °C for 3 h (pH) or 24 h (bile salts) and growth <10% at 0.3% bile (24 h)

(continued on next page)

79
80
Table 3 (continued)

Studies Materiels and methods Results


Mozzi et al. (2009) GS model (mouth and stomach CRL1190 and M16 Viable count by plating No significant loss: 8.6 log10 CFU/mL detected at pH 4.4 and 7.4 log10 CFU/mL at pH 3 for
model); 2.5 h digestion at 37 °C; CRL1190, 8.7 log10 CFU/mL at pH 4.4 and 8.0 log10 CFU/mL at pH 3 for M16
kinetics of gastric pH
Boke et al. (2010) pH tolerance 2, 2.5, 3 or 6.2; bile salts 2 high and 2 low EPS- Viable count by plating No viable cell detected at pH 2–2.5 and <7% survival with 0.15% and 0.3% bile salts for low
resistance: 0.15 or 0.3%; incubation at producing strains EPS-producing strains; 30–45% survival at pH 2 and 71% survival with 0.3% bile salts for
40 °C for 2 h high EPS-producing
Iyer et al. (2010) pH 2, 3 or 4; bile salts: 0.5, 1 or 2%. GI RD102 and RD104 Viable count by plating Few or no viability loss at different pH (2 h) and different bile concentrations (3 h) for
stresses: gastric juice pH 1.5, 2 or 3 both strains; few or no viability loss at pH 2 and 3 with or without pancreatic juice for
with or without pancreatic juice 0.5, both strains; complete loss of viability at pH 1.5 with or without the pancreatic juice for
1 or 2% of bile salts; incubation at both strains for GI stress conditions
37 °C for 2 h (pH); 3 h (bile salts); 3 h
(GI stresses)
Ziarno (2010) Simulated gastric juice: pH 2.4; 12 strains Viable count by plating Loss of 1.6–3.8 log10 CFU/mL after 5 h in gastric fluid; loss of 2.6–3.8 log10 CFU/mL after
simulated duodenal juice: 17 g/L bile; 6 h in duodenal fluid
incubation at 37 °C for 5 h (gastric) or
6 h (duodenum)
García-Hernández et al. Simulated gastric juice: pH 2; CECT801 Viable count by DVC-FISH 10.9% survival after 3 h in the simulated gastric juice and decrease of 15% of survival in

O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89


(2012) simulated pancreatic juice: pH 8; the simulated pancreatic juice after 6 h
incubation for 3 h or 6 h
Fang et al. (2013) Simulated gastric juice: pH 2, 2.5, 2.8 Cold- or non-shocked BCRC Viable count by plating No viable cell detected at pH 2 after 1 h and at pH 2.5 after 1–2 h for cold- or non-shocked
or 3; bile salts: 2%; incubation at 14085 strains; few or no viability loss at pH 2.8 and 3 and with 2% bile salts
37 °C for 4 h or 12 h
Ziar et al. (2014) Bile salts: 0, 2, 3, 4 g/L, 6 different TA040 Viable count by plating Loss of 0.5–1.5 log10 CFU/mL with 3 or 4 g/L bile; bacterial survival more affected in the
carbohydrates (5 g/L); incubation at presence of glucose (loss of 2 log10 CFU/mL at 4 g/L bile) than with lactose, raffinose,
37 °C for 12 h lactulose, pectin or lactulose
Junjua et al. (2016) pH 2 and 4; bile salts: 0.5 g/L of 30 strains DO600 nm measure for 20 h Comparison between non-stressed and stressed cell revealed great diversity of resistance
sodium cholate and 0.5 g/L of sodium (post-stress) to the GI tract stresses between strains
deoxycholate; incubation at 37 °C for
1h
Kebouchi et al. (2016) BSM: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 5, and LMD-9 Viable count by plating Significant loss of 5 log10 CFU/mL observed at 3 mM and no viable cell detected for
10 mM; incubation at 37 °C for 2 h concentration higher than 3 mM
Uriot et al. (2016) TIM model: dynamic stomach and 4 strains; 2 food matrices: Viable count by plating Loss of viability when gastric pH reached 2.5 at 45 min and global loss of 2–4 log10 CFU
small intestinal model; kinetics of non- and fermented milk for 3 tolerant strains; at the end of digestion, counts below 1 log10 CFU for the CNRZ21
gastric and intestinal pH; 5 h sensitive strain in all the digestive compartments; 6.1 and 8.5 log10 units for the sensitive
digestion at 37 °C and the 3 tolerant strains, resp., in the cumulative ileal effluents; higher survival in
fermented milk (1.6%) vs non-fermented milk (0.4%) for LMD-9

DVC: direct viable count; BSM: equimolar bile salt mixture containing sodium taurocholate, sodium cholate, and sodium deoxycholate; EPS: exopolysaccharide; FISH: fluorescent in situ hybridization; GF: germ-free; GI: gastro-
intestinal; GS: gastric system; HMA: human microbiota-associated; IBD: inflammatory bowel diseases; PCR; polymerase chain reaction; TIM: TNO gastrointestinal model; UCr: ulcerative colitis in remission; VSL-3: preparation of
probiotics.
O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89 81

Thomas et al., 2011). The results obtained in minipigs, rabbits and to acidic pH (pH 2 and 4), bile salts (0.5 g/L sodium cholate and
mice matched those found in humans: S. thermophilus survived 0.5 g/L sodium deoxycholate) and 10 mM H2O2 as well as their
through the GI tract and quickly disappeared from feces samples adhesion capacity to HT29-MTX intestinal epithelial cells. Adhe-
after cessation of consumption. The results obtained by Mater sion capacity is an important criterion for probiotic strains as it
et al. (2006) suggested that the disappearance of S. thermophilus may increase their ability to colonize the digestive tract. According
from the digestive tract may be due to competition with intestinal to Junjua et al. (2016), class 1 (8 strains) and 2 (5 strains) possess
microbiota. In their study, the survival of S. thermophilus FBI3 was low adhesion capacity and low resistance to bile salts and H2O2.
evaluated in germ-free (GF) and human-microbiota associated Members of class 3 (4 strains) and 4 (8 strains) resisted well to bile
(HMA) mice. When S. thermophilus was no longer administered, salts and H2O2 but had a low capacity of adhesion. The two strains
it disappeared faster from the small intestine of HMA-mice (6 h) of class 5 resisted well to acid stress and displayed the higher
than from that of GF-mice (10 h). Other studies in germ-free ani- adhesion capacities. The three strains that were able to resist to
mals have shown contradictory results. Drouault et al. (2002) con- pH 2 constituted the class 6.
cluded that S. thermophilus FBI3 was not able to settle in GF-mice Even if in vitro experiments in simple static systems bring valu-
while S. thermophilus LMD-9 and LMG18311 could colonized the able information about S. thermophilus sensitivity to specific
ileum and colon of GF-rats (Ben-Yahia et al., 2012; Thomas et al., parameters of digestion, they remain far from the complexity of
2011). In these latter studies, the colonization was improved by human digestive physiology. Complex dynamic in vitro models
lactose ingestion and the authors underlined the importance of which more closely reproduce the human digestive environment
glycolytic enzymes and lactose transporters in the ability of S. ther- can be alternatively used. The dynamic multi-compartmental
mophilus to adapt to the digestive environment and colonize the TNO gastrointestinal model (TIM) is currently one of the most
digestive tract of rats. complete simulator of the human stomach and small intestine
Taken together, these data show that in vivo studies in animals (Blanquet-Diot et al., 2012; Minekus, Marteau, Havenaar, & Huis
with defined microbiota provide helpful information, such as fac- in’t Veld, 1995). This model integrates the main parameters of
tors required to improve S. thermophilus survival or metabolic human digestion such as pH, body temperature, gastric and ileal
state. Nevertheless, the results obtained seem to vary depending deliveries, peristaltic mixing and transit, gastric, biliary and pan-
on the animal models and therefore should be confirmed before creatic secretions, passive absorption of water and digestion prod-
extrapolation to the human situation. ucts (Blanquet-Diot et al., 2012). Using this model, Marteau,
Minekus, Havenaar, and Huis in’t Veld (1997) showed that 12%
3.3. In vitro experiments of ingested S. thermophilus ST20 were able to reach the duodenum,
while less than 5% were recovered in the ileal effluents. Among the
For ethical, regulatory, technical and cost reasons, in vitro assays four LAB strains tested in that study (S. thermophilus, B. bifidum, Lb.
are advantageously used as an alternative to in vivo experiments. In acidophilus and Lb. bulgaricus), S. thermophilus was found to be the
vitro assays provide conditions for a better adjustment of selective more sensitive to GI passage (Marteau et al., 1997). In another
parameters of digestion, such as pH and bile salts concentrations, recent study using the same in vitro model, Uriot et al. (2016) con-
as well as a better understanding of their influence on bacterial firmed the low survival rate of S. thermophilus strains, especially in
survival and physiology. the small intestine, with recovery percentages in the ileal effluents
A large disparity of survival rates was observed when S. ther- less than 1%. Nevertheless, it is important to mention that bacte-
mophilus strains were subjected to pH conditions varying from rial amounts in the ileal effluents of the TIM ranged from 6 to
1.5 to 4 (Boke, Aslim, & Alp, 2010; Fang, Lai, & Chou, 2013; 8.5 CFU log10, which is supposed to be high enough to exert a pro-
García-Hernández et al., 2012; Iyer et al., 2010; Junjua et al., biotic activity (Minelli & Benini, 2008). These experiments in the
2016; Mozzi, Gerbino, Font de Valdez, & Torino, 2009; Vinderola TIM system also confirmed that the survival rates of S. ther-
& Reinheimer, 2003; Ziarno, 2010). However, most of strains were mophilus is highly strain-dependent: three of the tested strains
sensitive to pH values equal or lower than 3. Similarly, the resis- LMD-9, PB18O and EBLST20 showed significantly higher survival
tance of S. thermophilus to bile salts (bovine or porcine bile salts) capacities to GI conditions than the fourth one CNRZ21 (Uriot
was shown to be widely strain-dependant (Boke et al., 2010; et al., 2016). Analysis of multiple protein functions indicated that
Fang et al., 2013; García-Hernández et al., 2012; Iyer et al., 2010; GI low survival capacity could be attributed to the lack of heat
Junjua et al., 2016; Vinderola & Reinheimer, 2003) even if most shock protein and urease functions in CNRZ21. This study also
of strains were sensitive to bile salts concentrations higher than showed that the way how bacteria are administered has an impor-
0.5% w/v. Kebouchi et al. (2016) have further investigated the tant impact on their survival in the digestive tract. Indeed, S. ther-
effect of different concentrations of bile salts (0, 0.25, 0.5, 1, 1.5, mophilus LMD-9 survived better in gastric and small intestinal
2, 3, 4, 5 and 10 mM) including taurine-conjugated bile salts (tau- conditions when cells were administered as fermented milk
rocholate) and unconjugated bile salts (cholate and deoxycholate) (1.6%) rather than delivered as non-fermented milk (0.4%). Fer-
on S. thermophilus LMD-9 and showed that the strain was resistant mented milk seems therefore to be a good vehicle for S. ther-
to bile salts up to 3 mM (eq 1.5%). In addition, Ziar, Gérard, and mophilus strains.
Riazi (2014) and Boke et al. (2010) have shown that carbon sources Even if, like for animal models, results obtained using in vitro
and some bacterial factors may influence the ability of S. ther- models cannot be directly extrapolated to humans, these studies
mophilus to survive to pH and bile stresses. Bacterial survival was allow a better understanding of the impact of GI parameters, nutri-
more affected by bile salts in the presence of glucose than with lac- tional sources and mode of administration on the survival of S.
tose, mannitol, raffinose, pectin or lactulose. In particular, they thermophilus. In particular, dynamic multi-compartmental in vitro
showed that S. thermophilus TA040 displayed the same population digestion models, such as the TIM or the Simulator of the Human
level with 4 g/L bile or without bile when lactose was added to the Intestinal Microbial Ecosystem (SHIME) (Marzorati et al., 2014;
incubation medium (Ziar et al., 2014). Boke et al. (2010) also Molly, Vande Woestyne, & Verstraete, 1993) are useful to decipher
showed that two high exopolysaccharide (EPS)-producing S. ther- the behavior of probiotics in the successive environments of the
mophilus strains had a better resistance to acid pH and bile salts human GI tract and learn how they respond to the main physico-
than the two low EPS-producing strains. chemical digestive parameters (such as acid stress or bile but also
Interestingly, Junjua et al. (2016) have classified into six cate- oxygen level or fluid shear) in a physiological temporal-spatial
gories thirty strains of S. thermophilus according to their tolerance fashion.
82 O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89

4. Acid and bile resistance mechanisms in S. thermophilus As a conclusion, resistance strategies of S. thermophilus to acid
and bile are composed of a large combination of numerous differ-
Variability in GI survival capacity between S. thermophilus sug- ent mechanisms. This may explain the wide variability of tolerance
gests than some strains have developed specific resistance mecha- to GI stresses encountered among S. thermophilus strains.
nisms to the main stresses encountered in the digestive
environment, such as acid pH and bile.
5. Beneficial health effects of S. thermophilus
Responses of Gram positive bacteria to acidic stress are less doc-
umented that those of Gram negative bacteria. Cotter and Hill
A number of recent studies performed in humans or in animals
(2003) describe the acidity resistance of Gram positive bacteria as
have shown that S. thermophilus may have beneficial effects on
‘‘a combination of constitutive and inducible strategies which result
host health, as described below.
in the removal of protons (H+), alkalization of the external environment,
changes in the composition of the cell envelope, production of general
shock proteins and chaperones, expression of transcriptional regulators, 5.1. Alleviation of lactose intolerance
and responses to changes in cell density”. Few data are available on the
mechanisms of acid resistance for S. thermophilus. Arena et al. Lactose intolerance is the inability of adults and children to
(2006) suggested that the response to acidic stress of S. thermophilus digest the milk sugar lactose. This is due to a deficiency of the
combines pH homeostasis through over-expression of H+-ATPase, enzyme b-galactosidase in the brush border of the small intestine.
reduction of intracellular lactic acid by the up-regulation of lactate When the b-galactosidase is present, lactose is hydrolyzed into its
dehydrogenase and increase in ammonium concentration by the constituent monosaccharides, glucose and galactose, which are
up-regulation of urease. In accordance with these data, Uriot et al. transported across the small intestine epithelium. Individuals with
(2016) found that S. thermophilus CNRZ21 which is devoid of the lactose intolerance suffer from excessive flatulence, abdominal
urease activity had a much lower survival capacity in the TIM sys- pains and diarrheas. In 2010, EFSA has granted a health claim to
tem than the three other strains which express the enzyme. Never- yogurt for improvement of lactose digestion. The claim was formu-
theless, the role of urease in S. thermophilus acid tolerance remains lated as follows: ‘‘Live yogurt cultures in yogurt improve digestion of
controversial. Even if most of the 30 S. thermophilus strains tested in lactose in yogurt in individuals with lactose maldigestion” (EFSA,
Junjua’s study possess the urease function, the authors indicates 2010). This claim is based on 14 human intervention studies. These
that urease does not seem to be a significant factor in acidic stress studies have shown an enhanced digestion of lactose by persons
tolerance. In addition, Zotta, Ricciardi, Ciocia, Rossano, and with lactose intolerance when consuming fresh yogurt compared
Parente (2008) and Arioli et al. (2010) showed that the urease was to pasteurized yogurt with reduced or no live bacteria. To bear
not very active at low acidic pH, closed to that found in the human the claim, yogurt must contain at least 108 CFU live microorgan-
stomach. They suggested that urease would rather have a metabolic isms of Lb. bulgaricus and S. thermophilus per gram of fermented
role by optimizing the activity of S. thermophilus glycolytic enzymes product. The claim has been attributed to yogurt containing both
(Arioli et al., 2010; Zotta, Ricciardi, Rossano, & Parente, 2008). bacteria, but not to S. thermophilus alone. In order to better under-
Other factors have been proposed to be involved in acid resis- stand the specific role of S. thermophilus in lactose intolerance, sev-
tance, as they were found to be over-expressed under low pH: eral studies have been performed in gnotobiotic animal models. In
the chaperone GroEL and GroES which are involved in protein GF mice, orally administrated S. thermophilus cells were able to
repair, acidic shock proteins (Asp or Hsp in the literature), peroxide produce an active b-galactosidase in the GI tract (Drouault et al.,
resistance protein Dpr, general stress protein 24, translation elon- 2002). When these mice received lactose (4.5% wt/vol) as drinking
gation factor (EF) G, Tu and Ts, phosphoglycerate mutase and cell water, a significant diminution of lactose in feces was observed
division protein DivIVA (Arena et al., 2006; González-Márquez, compared to control mice without S. thermophilus cells. Studies
Perrin, Bracquart, Guimont, & Linden, 1997; Zotta et al., 2008). using HMA-mice showed that b-galactosidase production by S.
The role of Hsp in acid tolerance is strengthened by the study of thermophilus occurred in the second half of the small intestine
Tian et al. (2012) who showed that under acid condition (pH 3), but not in the colon (Mater et al., 2006).
the survival of L. lactis ML23 was improved when cells were har- Then, even if the beneficial effect of yogurt on alleviation of lac-
boring a plasmid carrying the shsp gene of S. thermophilus St-QC. tose intolerance is well established by clinical studies, there are
When combining the data obtained by Uriot et al. (2016) and strong proofs in animals but yet no solid evidence in humans that
Junjua et al. (2016), it can be observed an obvious correlation this effect can be specifically attributed to S. thermophilus.
between Hsp and acid tolerance: the three S. thermophilus strains
that are able to survive under pH 2 possess Hsp (LMD-9, PB180 5.2. Prevention of chronic gastritis
and PB302) and 16 out of the 20 most resistant strains to pH 4 also
possess Hsp. Lastly, amino acid decarboxylases (Trip, Mulder, & Studies on animal models have shown that S. thermophilus
Lolkema, 2012) and antioxidant pathways such as manganese- could prevent the development of gastritis, an inflammatory dis-
dependent superoxide dismutase (MnSOD, sodA gene) (Bruno- ease of the stomach that can be caused by the intensive use of
Bárcena, Azcárate-Peril, & Hassan, 2010) may also have a role in acetylsalicylic acid (ASA). In the study by Rodríguez, Medici,
the acid resistance of S. thermophilus. Several of these proteins Rodríguez, Mozzi, and Font de Valdez (2009), administration of a
involved in stress response (such as SodA, GroEL, EF-Tu) were also daily dose of milk fermented by S. thermophilus CRL1190 (108 -
found to be overproduced in feces of rats mono-associated with S. CFU/day) given for seven days was found to protect mice against
thermophilus LMD-9 compared to milk growth (Rul et al., 2011). gastritis induced by the administration of ASA. The gastro-
Bile resistance mechanisms of S. thermophilus are poorly stud- protective effect was clearly attributed to the EPS produced by
ied. As for acidity, production of Hsp could be used by S. ther- the S. thermophilus strain. EPS could stimulate the immune system
mophilus to resist to the deleterious effect of bile salts (Tian and exert an inhibitory effect on ulcer in the host (Rodríguez et al.,
et al., 2012). Besides, it has been recently shown that two 2009). Further studies conducted by the same authors (Rodríguez,
sortase-dependant proteins, PrtS and MucBP, found in the bacterial Medici, Mozzi, & Font de Valdez, 2010) indicated that the EPS pro-
cell surface allow a better survival in presence of bile salts duced by S. thermophilus CRL1190 had the same effect than the
(Kebouchi et al., 2016). Omeprazole drug used for the treatment of gastritis.
O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89 83

As animal studies showed very promising results regarding the genes and Bacillus cereus. Production of thermophilins represents a
use of a specific S. thermophilus strain in the prevention of chronic significant interest for conservation in food industry because it
gastritis, it would be interesting to investigate such beneficial may limit the proliferation of pathogenic bacteria in dairy products
effects in humans. A natural therapy using purified EPS or milk fer- fermented by S. thermophilus (Rossi et al., 2013).
mented by S. thermophilus CRL1190 would be a relevant alternative In the context of infection with C. difficile, mice treated with
to proton-pump inhibitors, such as Omeprazole, whose use is viable S. thermophilus exhibited 46% less weight loss compared
restricted by undesirable side effects and drug interactions. with untreated controls (Kolling et al., 2012). The treatment also
led to lower diarrheal severity, lower pathology scores and
decrease in toxin production. An inverse correlation between the
5.3. Prevention of diarrhea
levels of luminal lactic acid and abundance of C. difficile was also
observed, suggesting that lactic acid produced by S. thermophilus
Acute diarrhea is a serious cause of infant mortality that can
may impact pathogen growth in the murine system.
result from viral (rotavirus) or bacterial infections in the gut.
Antimicrobial activity is a main trait of probiotic properties. S.
Saavedra, Bauman, Oung, Perman, and Yolken (1994) showed that
thermophilus is able to produce thermophilins, but as a part of pro-
formula combining B. bifidum and S. thermophilus reduced the inci-
biotic criteria, it is necessary to test whether these bacteriocins are
dence of rotavirus-associated diarrhea in infant aged 5–24 months
produced in the GI tract and are effective against pathogenic bac-
from 31% (control, n = 26) to 7% (treated group, n = 29). In another
teria in such environment. S. thermophilus also produces lactic acid,
study from the same group (Saavedra, Abi-Hanna, Moore, & Yolken,
another antimicrobial compound, but this trait, shared by all LAB,
2004), healthy infant aged 3–24 months supplemented with live B.
is not specific to this bacterium. Again, it would be interesting to
lactis and S. thermophilus were less susceptible to develop colitis,
evaluate the impact of lactic acid production by S. thermophilus
which consequently reduced the use of antibiotics. In a large study
on its capacity to colonize the GI tract and compete against food-
involving 571 children aged 3–36 months and consulting for acute
borne pathogens.
diarrhea, administration of five different probiotic preparations
were tested in their effectiveness in reducing diarrhea duration.
The probiotic bacterial mixture including S. thermophilus gave sig-
nificantly shorter diarrhea (70 h) than oral rehydration solution
6.2. Antioxidant activity
alone (115 h) (Canani et al., 2007).
Antibiotic-associated diarrhea (AAD) is a common adverse
Some promising antioxidant activities of S. thermophilus have
effect of antibiotic therapy. Scarce studies have investigated the
been observed in both in vitro and in vivo models. Among 49 strains
effectiveness of yogurt consumption in the prevention of AAD. In
of LAB, S. thermophilus YIT2001 showed the highest inhibitory
the randomized study of Beniwal et al. (2003), adult patients
activity against lipid peroxidation in liposomes treated by ferrous
receiving 227 g of yogurt per day for 8 days developed less diar-
iron (Ito, Ohishi, Yoshida, Yokoi, & Sawada, 2003). Feeding iron-
rhea (12.4%, n = 105) than control (23.7%, n = 97). On the opposite,
overloaded mice for 2 weeks with S. thermophilus YIT2001 caused
another randomized study on 369 patients failed to demonstrate
a decrease in lipid peroxide in the colonic mucosa of the animals.
that yogurt had a positive effect on AAD both in children and in
The authors suggested that S. thermophilus YIT2001 may act as a
adults (Conway, Hart, Clark, & Harvey, 2007). These contradictory
scavenger of reactive oxygen species or free radicals or may
results may be mostly due to patient complexity but also to the
increase antioxidant capacities of intestinal content, but seemed
type of yogurt and ingested dose. In a recent meta-analysis by
not to work through removal of iron (Ito et al., 2003). This strain
Patro-Golab, Shamir, and Szajewska (2015), the authors concluded
was also able to prevent oxidative injuries to the colonic mucosa
that there is insufficient evidence to support or refute the use of
and improve the disease activity index in dextran sulfate sodium
yogurt in preventing AAD, given the small number of trials and
(DSS) induced colitis mice, as well as reduce associated anemia
participants as well as the methodological limitations of the
(Ito et al., 2008). In a last study by the same authors (Ito et al.,
included trials (Patro-Golab et al., 2015).
2015), S. thermophilus YIT2001 showed the ability to inhibit low
To conclude, up to date, there are some evidences that yogurt
density lipoprotein (LDL) oxidation and reduce aortic fatty lesions
consumption can reduce the incidence of infectious diarrhea,
in hyperlipidemic hamsters. S. thermophilus 1131 also had an
which is not clearly demonstrated in AAD. In any case, the benefi-
inhibiting effect on LDL oxidation in blood samples collected from
cial role of S. thermophilus in preventing diarrhea is not yet fully
one human volunteer (Terahara, Kurama, & Takemoto, 2001). The
established and should be further analyzed.
underlying mechanisms of such antioxidant effects are not yet
known but could be related to the activity of antioxidant enzymes
6. Possible mechanisms of action of S. thermophilus on the host produced by S. thermophilus, like SOD or glutathione reductase
health (Amaretti et al., 2013; Chang & Hassan, 1997). By using menadione
(a superoxide-generating agent)-sensitive mutants, Thibessard
6.1. Antimicrobial activity et al. (2004) have characterized also three genes (tgt, ossF and ossG)
in S. thermophilus CNRZ368 whose action might contribute to
S. thermophilus is able to synthesize thermophilins which are oxidative stress defense.
bacteriocins, small peptides able to inhibit the growth or kill clo- Taken together, these studies indicate that some S. thermophilus
sely related bacteria (Dortu & Thonart, 2009). Hitherto, ten ther- strains may have a beneficial antioxidant effect both in vitro and
mophilins have been identified from ten different strains of S. in vivo in animals. Nevertheless, it should be underlined that most
thermophilus (Table 4). Thermophilins have been shown to have of available studies involve a single strain of S. thermophilus
in vitro inhibitory activities against LAB but also against Gram pos- YIT2001. It seems that several antioxidant mechanisms are
itive pathogenic strains such as E. faecalis, Clostridium (C.) botuli- involved and knowledge of a more exhaustive panel of the bacte-
num, Staphylococcus aureus and Listeria monocytogenes (Fontaine rial potential molecules would facilitate the selection of super
& Hols, 2008; Rossi, Marzotto, Cremonese, Rizzotti, & Torriani, anti-oxydant strains. These results open up new opportunities for
2013). For example, thermophilin 1277, which is produced by S. the use of S. thermophilus in the prevention of various diseases
thermophilus SBT1277, shows an antimicrobial activity against sev- where oxidative stress has been shown to play a key role, such
eral LAB and food spoilage bacteria including C. butyricum, C. sporo- as ulcerative colitis, colon cancer or artherosclerosis.
84
Table 4
Characterization of some bacteriocins produced by S. thermophilus.

Thermophilins 110 1277 13 347 580 81 9 A ST-1 T


S. thermophilus ST110 SBT1277 SFi13 347 580 81 LMD-9 ST134 ACA-DC0001 ACA-DC 0040
producing strains
References Gilbreth and Kabuki, Uenishi, Marciset, Villani et al. (1995) Mathot, Ivanova et al. Fontaine and Ward and Somkuti Aktypis and Aktypis,
Somkuti (2005) Watanabe, Seto, Jeronimus- Beliard, and (1998) Hols (2008) (1995) Kalantzopoulos Kalantzopoulos,
and Nakajima Stratingh, Mollet, Thuault (2003) Huis in’t Veld, and
(2007) and Poolman (2003) ten Brink (1998)
(1997)

O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89


Classification ND Lantibiotic Class IIa Class IIa ND ND Class IIa Class IIa Class IV Class II
MW (Da) 4000–4500 3700 ND 2500–6200 ND 4500 ND 1700 30000 2500
Enzyme sensitivity Protease Proteinase K ND a-chymotrypsin, Protease and Proteinase K ND Protease sensitive Pronase and Protease and a-
sensitive and trypsin trypsin, protease K a-amylase and pronase E Amyloglucosidase, trypsin amylase sensitive
Inactivated by sensitive sensitive sensitive sensitive lysozyme and b- sensitive Lysozyme and
a-amylase Pepsin resistant Catalase and amylase resistant lipase resistant
lipase
resistant
Heat sensitivity Stable after 1 h Activity ND Stable after 1 h at Not heat- Not heat- ND Stable after 1 h at Not heat- Inactive after
at 100 °C maintained 100 °C. 50% of resistant resistant 100 °C resistant 20 min at 121 °C
after 1 h at activity loss after Inactive after Inactive after Inactive after
130 °C 15 min at 121 °C 1 h at 60 °C 15 min at 10 min at 60 °C
60 °C
pH tolerance ND Active between ND ND ND Active ND Active between Active between Active between
pH3 and pH10 between pH3 pH3 and pH7 pH3 and pH10 pH1 and pH9
and pH10
Antimicrobial activities E. faecalis C. butylicum B. cereus E. faecalis B. cereus B. cereus L. S. thermophilus S. aureus LAB
/ strains L. lactis C. sprogenes C. botulinum L. lactis C. butyricum B. subtilis monocytogenes L. innocua C. sporogenes
Lb. bulgaricus Lb. acidophilus L. monocytogenes L. monocytogenes C. sporogenes E. coli S. thermophilus E. faecalis C. tyrobutyricum
P. acidilactici Lb. helveticus LAB S. thermophilus E. faecalis E. faecalis
S. thermophilus M. lacticum S. thermophilus Lb.
S. thermophilus monocytogenes
S. typhimurium
No antimicrobial S. aureus L. innocua E. coli C. sporogenes E. coli LAB C. sporogenes E. faecalis
activity/strains S. epidermidis L. Pseudomonas E. coli L. lactis P. damnosus C. tyrobutyrium L. innocua
monocytogenes Salmonella Lb. bulgaricus L. S. aureus S. carnosus
Lb. gasseri Lb. helveticus monocytogenes
S. aureus S. aureus Lb. helveticus
S. aureus

HT: heat treatment; LAB: lactic acid bacteria; MW: molecular weight; ND: not determined.
O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89 85

6.3. Immunomodulation and Lb. acidophilus ATCC4356 prevents decrease in trans-epithelial


resistance induced by entero-invasive Escherichia coli (EIEC). The
A number of in vitro studies have investigated the ability of S. effect of the two bacteria on resistance was found to be accompa-
thermophilus strains to modulate the immune response of various nied by maintenance (actin, ZO-1) or enhancement (actinin,
human cell lines, such as intestinal HT-29 cells, Peripheral Blood occluding) of cytoskeletal and tight junctional protein phosphory-
Mononuclear Cells (PBMCs), monocyte-derived Dendritic Cells lation. S. thermophilus and Lb. acidophilus also enhanced the barrier
(moDCs) or human primary macrophages. Depending on the study function of naive epithelial cells not exposed to any pathogen.
and strain tested, either pro-inflammatory or anti-inflammatory Apart from their effect on barrier function, the authors showed that
responses were obtained. Kekkonen et al. (2008) found that S. ther- pretreatment with the two strains limited the number of adhered
mophilus THS induced Th1 type cytokines Interleukin (IL)-12 and and invasive EIEC. The ability of S. thermophilus to improve intesti-
Interferon-gamma (IFN-c) as well as Tumor Necrosis Factor- nal barrier function was confirmed by an in vivo study in 35
alpha (TNF-a) in PMBCs. These results are in accordance with those healthy human subjects (Del Piano et al., 2014). Supplementation
of Latvala et al. (2008) who showed that the same strain induced with S. thermophilus ST10 and tara gum was able to significantly
the expression of pro-inflammatory (TNF-a, IL-12, IL-6, Chemokine decrease intestinal permeability, both in the small bowel and in
CC Ligand -CCL- 20) and Th1 type (IL-12 and IFN-c) cytokines in the colon of the volunteers. The authors observed a parallel
moDCs. The same authors confirmed in a more recent study increase in EPS concentration in the fecal material. When embed-
(Latvala, Miettinen, Kekkonen, Korpela, & Julkunen, 2011) that S. ded in tara gum, EPS would act by forming a film over the inner
thermophilus THS is a strong inducer of IL-12 in moDCs, PBMCs surface of the bowel, therefore creating a mechanical barrier.
and macrophages. On the contrary, Latvala et al. (2011) showed So far, scarce studies have shown the potential of S. ther-
that S. thermophilus is a potent inducer of the anti-inflammatory mophilus in improving epithelial barrier function and should be
IL-10 in macrophages. Such anti-inflammatory effect was con- further deepened to identify underlying mechanisms. If the benefi-
firmed by the study of Junjua et al. (2016). Among 30 S. ther- cial properties of S. thermophilus are confirmed by further in vivo
mophilus strains of different origins, they found that most strains studies in humans, it opens up new prospects for using this bac-
reduced the production of pro-inflammatory IL-8 after co- terium in the management of many pathologies.
incubation with HT-29 cells, while they induced the production
of IL-10 in PBMC. The authors calculated the ration of synthesis
6.5. Interactions with intestinal microbiota
IL-10/IL-12 and showed that three strains (CNRZ21, CNRZ160 and
PB5MJ) displayed a strong and promising in vitro anti-
There is no available study investigating the interactions
inflammatory potential with a ratio higher than 100. Such a value
between S. thermophilus and the human intestinal microbiota. Nev-
is similar or higher than the one reported for Faecalibacterium
ertheless, García-Albiach et al. (2008) have evaluated the effect of
prausnitzii, a commensal bacteria displaying one of the best anti-
yogurt on the gut microbiota of 79 healthy young adults. The main
inflammatory properties (Sokol et al., 2008). Ogita et al. (2011)
change observed after yogurt consumption was an increase in the
suggested that modulation of Th1/Th17 balance would be one of
levels of LAB and C. perfringens to the detriment of Bacteroides.
the mechanisms used by S. thermophilus ST28 to exert an anti-
Then, even if it is generally acknowledged that probiotics act
inflammatory effect. They showed that heat-killed S. thermophilus
through beneficial modulation of gut microbiota, much remains
ST28 suppressed IL-17 production in murine splenocytes stimu-
to be done in that field.
lated with Transforming Growth Factor-beta (TGF-b) plus IL-6
(Ogita, Tanii, Morita, Suzuki, & Tanabe, 2011). This in vitro effect
was confirmed in vivo in DSS-treated mice where oral treatment 7. Conclusion
by the strain decreased the production of IL-17 and the percent-
ages of Th17 cells in lamina propria lymphocytes (Ogita, Naka- Critical criteria to be recognized as a probiotic strain which can
shima, et al., 2011). Lastly, in a study designed to follow a large be used for manufacturing of functional foods are survival in GI
number of cytokines (IL-2, IL-4, IL-6, IL-10, IL-12, IL-17, IFN-c, conditions, non-pathogenicity, proved beneficial health effects
TGF-b), Donkor et al. (2012) concluded that S. thermophilus and resistance to industrial process (Miquel et al., 2015; Nagpal
St1275 induced significant secretion of both pro- and anti- et al., 2012; Vasiljevic & Shah, 2008).
inflammatory cytokines from PBMC. Interestingly, Del Carmen Even if most of S. thermophilus strains appeared to be sensitive
et al. (2014) have observed that introducing an antioxidant func- to acid pH and bile salts, human studies have established their abil-
tions (catalase and SOD) enhances the anti-inflammatory activity ity to survive passage through the GI tract and transiently colonize
of S. thermophilus strains. while ingested. In addition, some strains of S. thermophilus have the
Hitherto, there is no consensus in the literature about the ability to adhere to intestinal epithelial cells, which is an important
immunomodulatory properties of S. thermophilus that seem to be criterion for probiotic strain selection. This biological mechanism
widely dependent on both the strain tested and the type of cells promoting interactions with the host leads to gut protection and
used. If the anti-inflammatory properties of some specific strains enhances colonization. With regards to safety, S. thermophilus is
of S. thermophilus are further demonstrated, they could be involved already considered as safe by the FDA. A large number of in vivo
in the development of novel therapeutic products that prevent studies in human or animal models have also shown beneficial
pathologies where inflammation plays a key role, such as inflam- health effects for S. thermophilus, such as alleviation of lactose
matory bowel diseases. intolerance, prevention of gastritis and prevention of infectious
diarrhea. The mode of action of S. thermophilus has been already
6.4. Effect on intestinal barrier function investigated, by using in vitro assays and animal models. The bac-
terium seems to act mainly through the production of antimicro-
Hyper-permeability of the intestinal barrier has been associated bial compounds (such as thermophilins), but also via its
with a variety of pathologic states like infection, celiac disease, antioxidant and anti-inflammatory properties or its ability to
Crohn’s disease or type 1 diabete. Use of probiotic bacteria raises enhance epithelial barrier function. Nevertheless, the available
hope in restoring impaired barrier function. Resta-Lenert and data should be interpreted with caution as in a substantial part
Barrett (2003) showed that exposure of intestinal cell monolayers of these studies S. thermophilus was administrated with another
(both HT29 and caco-2 cell lines) to live S. thermophilus ATCC19258 LAB strain or within yogurt. In addition, even if some assumptions
86 O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89

have been made about mechanisms of action of S. thermophilus, Bouhafs, L., Moudilou, E. N., Exbrayat, J. M., Lahouel, M., & Idoui, T. (2015). Protective
effects of probiotic Lactobacillus plantarum BJ0021 on liver and kidney
they need to be confirmed in a larger number of in vivo studies,
oxidative stress and apoptosis induced by endosulfan in pregnant rats. Renal
first in animals then in human trials. In particular, there is almost Failure, 37(8), 1370–1378. http://dx.doi.org/10.3109/0886022X.2015.1073543.
no information about interactions of the bacteria with human gut Bowen, J. M., Stringer, A. M., Gibson, R. J., Yeoh, A. S. J., Hannam, S., & Keefe, D. M. K.
microbiota, even if microbiota modulation is a generally acknowl- (2007). VSL#3 probiotic treatment reduces chemotherapy-induced diarrhea
and weight loss. Cancer Biology & Therapy, 6(9), 1449–1454.
edged mode of action for probiotic strains. Knowing that both the Boyle, R. J., & Tang, M. L. K. (2006). The role of probiotics in the management of
survival and beneficial effects of S. thermophilus are widely strain- allergic disease. Clinical and Experimental Allergy: Journal of the British Society for
dependent, it is expected that only certain strains could obtain the Allergy and Clinical Immunology, 36(5), 568–576. http://dx.doi.org/10.1111/
j.1365-2222.2006.02472.x.
probiotic status, after clinical studies. Lastly, the resistance of S. Brigidi, P., Swennen, E., Vitali, B., Rossi, M., & Matteuzzi, D. (2003). PCR detection of
thermophilus to industrial process is already well established, Bifidobacterium strains and Streptococcus thermophilus in feces of human
mainly in dairy products. This represents a strong advantage for subjects after oral bacteriotherapy and yogurt consumption. International
Journal of Food Microbiology, 81(3), 203–209.
designing new probiotic dairy food, especially fermented milks, Bruno-Bárcena, J. M., Azcárate-Peril, M. A., & Hassan, H. M. (2010). Role of
using the appropriate strains. Indeed, fermented milks and yogurts antioxidant enzymes in bacterial resistance to organic acids. Applied and
benefits from favorable public opinion and are highly consumed Environmental Microbiology, 76(9), 2747–2753. http://dx.doi.org/10.1128/
AEM.02718-09.
(about 2 kg of yogurt per person worldwide), which should facili- Campus, G., Cocco, F., Carta, G., Cagetti, M. G., Simark-Mattson, C., Strohmenger, L., &
tate the commercialization of new products of this type. In addi- Lingström, P. (2014). Effect of a daily dose of Lactobacillus brevis CD2 lozenges
tion, the number of alive bacterial cells which reach the colon in high caries risk schoolchildren. Clinical Oral Investigations, 18(2), 555–561.
http://dx.doi.org/10.1007/s00784-013-0980-9.
can be increased by improving bacteria survival in yogurt, as it
Canani, R. B., Cirillo, P., Terrin, G., Cesarano, L., Spagnuolo, M. I., De Vincenzo, A., ...
was shown after the addition of the glucose oxidase to this food Guarino, A. (2007). Probiotics for treatment of acute diarrhoea in children:
(Batista et al., 2015). Randomised clinical trial of five different preparations. BMJ (Clinical Research
Then, this review has presented numerous studies which show Ed.), 335(7615), 340. http://dx.doi.org/10.1136/bmj.39272.581736.55.
Chang, S. K., & Hassan, H. M. (1997). Characterization of superoxide dismutase in
the potential of S. thermophilus as a new promising probiotic Streptococcus thermophilus. Applied and Environmental Microbiology, 63(9),
candidate. 3732–3735.
Conway, S., Hart, A., Clark, A., & Harvey, I. (2007). Does eating yogurt prevent
antibiotic-associated diarrhoea? A placebo-controlled randomised controlled
References trial in general practice. The British Journal of General Practice: The Journal of the
Royal College of General Practitioners, 57(545), 953–959. http://dx.doi.org/
10.3399/096016407782604811.
Aktypis, A., & Kalantzopoulos, G. (2003). Purification and characterization of
Cotter, P. D., & Hill, C. (2003). Surviving the acid test: Responses of gram-positive
thermophilin ST-1, a novel bacteriocin produced by Streptococcus thermophilus
bacteria to low pH. Microbiology and Molecular Biology Reviews, 67(3), 429–453.
ACA-DC 0001. Le Lait, 83(5), 365–378. http://dx.doi.org/10.1051/lait:2003024.
http://dx.doi.org/10.1128/MMBR.67.3.429-453.2003.
Aktypis, A., Kalantzopoulos, G., Huis in’t Veld, J. H., & ten Brink, B. (1998).
del Campo, R., Bravo, D., Cantón, R., Ruiz-Garbajosa, P., García-Albiach, R., Montesi-
Purification and characterization of thermophilin T, a novel bacteriocin
Libois, A., ... Boquero, F. (2005). Scarce evidence of yogurt lactic acid bacteria in
produced by Streptococcus thermophilus ACA-DC 0040. Journal of Applied
human feces after daily yogurt consumption by healthy volunteers. Applied and
Microbiology, 84(4), 568–576.
Environmental Microbiology, 71(1), 547–549. http://dx.doi.org/10.1128/
Amaretti, A., di Nunzio, M., Pompei, A., Raimondi, S., Rossi, M., & Bordoni, A. (2013).
AEM.71.1.547-549.2005.
Antioxidant properties of potentially probiotic bacteria: in vitro and in vivo
Del Carmen, S., de Moreno de LeBlanc, A., Martin, R., Chain, F., Langella, P.,
activities. Applied Microbiology and Biotechnology, 97(2), 809–817. http://dx.doi.
Bermúdez-Humarán, L. G., & LeBlanc, J. G. (2014). Genetically engineered
org/10.1007/s00253-012-4241-7.
immunomodulatory Streptococcus thermophilus strains producing antioxidant
Arena, S., D’Ambrosio, C., Renzone, G., Rullo, R., Ledda, L., Vitale, F., ... Scaloni, A.
enzymes exhibit enhanced anti-inflammatory activities. Applied and
(2006). A study of Streptococcus thermophilus proteome by integrated analytical
Environmental Microbiology, 80(3), 869–877. http://dx.doi.org/10.1128/
procedures and differential expression investigations. Proteomics, 6(1),
AEM.03296-13.
181–192. http://dx.doi.org/10.1002/pmic.200402109.
Del Piano, M., Balzarini, M., Carmagnola, S., Pagliarulo, M., Tari, R., Nicola, S., ... Pane,
Arioli, S., Ragg, E., Scaglioni, L., Fessas, D., Signorelli, M., Karp, M., ... Mora, D. (2010).
M. (2014). Assessment of the capability of a gelling complex made of tara gum
Alkalizing reactions streamline cellular metabolism in acidogenic
and the exopolysaccharides produced by the microorganism Streptococcus
microorganisms. PLoS ONE, 5(11), e15520. http://dx.doi.org/10.1371/journal.
thermophilus ST10 to prospectively restore the gut physiological barrier: A pilot
pone.0015520.
study. Journal of Clinical Gastroenterology, 48(Suppl 1), S56–61. http://dx.doi.org/
Bai, Y., Sun, E., Shi, Y., Jiang, Y., Chen, Y., Liu, S., ... Ren, F. (2016). Complete genome
10.1097/MCG.0000000000000254.
sequence of Streptococcus thermophilus MN-BM-A01, a strain with high
Delorme, C., Bartholini, C., Bolotine, A., Ehrlich, S. D., & Renault, P. (2010).
exopolysaccharides production. Journal of Biotechnology, 224, 45–46. http://dx.
Emergence of a cell wall protease in the Streptococcus thermophilus
doi.org/10.1016/j.jbiotec.2016.03.003.
population. Applied and Environmental Microbiology, 76(2), 451–460. http://dx.
Ballesta, S., Velasco, C., Borobio, M. V., Argüelles, F., & Perea, E. J. (2008). Fresh versus
doi.org/10.1128/AEM.01018-09.
pasteurized yogurt: Comparative study of the effects on microbiological and
Di Cerbo, A., Palmieri, B., Aponte, M., Morales-Medina, J. C., & Iannitti, T. (2016).
immunological parameters, and gastrointestinal comfort. Enfermedades
Mechanisms and therapeutic effectiveness of lactobacilli. Journal of Clinical
Infecciosas y Microbiología Clínica, 26(9), 552–557.
Pathology, 69(3), 187–203. http://dx.doi.org/10.1136/jclinpath-2015-202976.
Batista, A. L. D., Silva, R., Cappato, L. P., Almada, C. N., Garcia, R. K. A., Silva, M. C., ...
Dilmi-Bouras, A., & Sadoun, D. (2002). Survie des ferments du yaourt dans le tube
Cruz, A. G. (2015). Quality parameters of probiotic yogurt added to glucose
digestif du lapin. Le Lait, 82(2), 247–253. http://dx.doi.org/10.1051/
oxidase compared to commercial products through microbiological, physical-
lait:2002007.
chemical and metabolic activity analyses. Food Research International, 77,
Donkor, O. N., Ravikumar, M., Proudfoot, O., Day, S. L., Apostolopoulos, V., Paukovics,
627–635.
G., ... Gill, H. (2012). Cytokine profile and induction of T helper type 17 and
Beniwal, R. S., Arena, V. C., Thomas, L., Narla, S., Imperiale, T. F., Chaudhry, R. A., &
regulatory T cells by human peripheral mononuclear cells after microbial
Ahmad, U. A. (2003). A randomized trial of yogurt for prevention of antibiotic-
exposure. Clinical and Experimental Immunology, 167(2), 282–295. http://dx.doi.
associated diarrhea. Digestive Diseases and Sciences, 48(10), 2077–2082.
org/10.1111/j.1365-2249.2011.04496.x.
Ben-Yahia, L., Mayeur, C., Rul, F., & Thomas, M. (2012). Growth advantage of
Dortu, C., & Thonart, P. (2009). Les bactériocines des bactéries lactiques:
Streptococcus thermophilus over Lactobacillus bulgaricus in vitro and in the
Caractéristiques et intérêts pour la bioconservation des produits alimentaires.
gastrointestinal tract of gnotobiotic rats. Beneficial Microbes, 3(3), 211–219.
Biotechnologie, Agronomie, Societe et Environnement, 13(1), 143–154.
http://dx.doi.org/10.3920/BM2012.0012.
Drouault, S., Anba, J., & Corthier, G. (2002). Streptococcus thermophilus is able to
Blanquet-Diot, S., Denis, S., Chalancon, S., Chaira, F., Cardot, J.-M., & Alric, M. (2012).
produce a -galactosidase active during its transit in the digestive tract of germ-
Use of artificial digestive systems to investigate the biopharmaceutical factors
free mice. Applied and Environmental Microbiology, 68(2), 938–941. http://dx.
influencing the survival of probiotic yeast during gastrointestinal transit in
doi.org/10.1128/AEM.68.2.938-941.2002.
humans. Pharmaceutical Research, 29(6), 1444–1453. http://dx.doi.org/10.1007/
EFSA (2010). Scientific opinion on the substantiation of health claims related to live
s11095-011-0620-5.
yoghurt cultures and improved lactose digestion (ID 1143, 2976) pursuant to
Boke, H., Aslim, B., & Alp, G. (2010). The role of resistance to bile salts and acid
Article 13(1) of Regulation (EC) No 1924/2006. EFSA Journal, 8(10), 1763.
tolerance of exopolysaccharides (EPSS) produced by yogurt starter bacteria.
Elli, M., Callegari, M. L., Ferrari, S., Bessi, E., Cattivelli, D., Soldi, S., ... Antoine, J.-M. .
Archives of Biological Sciences, 62(2), 323–328. http://dx.doi.org/10.2298/
(2006). Survival of yogurt bacteria in the human gut. Applied and Environmental
ABS1002323B.
Microbiology, 72(7), 5113–5117. http://dx.doi.org/10.1128/AEM.02950-05.
Bolotin, A., Quinquis, B., Renault, P., Sorokin, A., Ehrlich, S. D., Kulakauskas, S., ...
Facklam, R. (2002). What happened to the streptococci: Overview of taxonomic and
Hols, P. (2004). Complete sequence and comparative genome analysis of the
nomenclature changes. Clinical Microbiology Reviews, 15(4), 613–630. http://dx.
dairy bacterium Streptococcus thermophilus. Nature Biotechnology, 22(12),
doi.org/10.1128/CMR.15.4.613-630.2002.
1554–1558. http://dx.doi.org/10.1038/nbt1034.
O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89 87

Fang, S.-H., Lai, Y.-J., & Chou, C.-C. (2013). The susceptibility of Streptococcus (Oslo, Norway: 1992), 98(1), 127–131. http://dx.doi.org/10.1111/j.1651-
thermophilus 14085 to organic acid, simulated gastric juice, bile salt and 2227.2008.00977.x.
disinfectant as influenced by cold shock treatment. Food Microbiology, 33(1), Ito, M., Ohishi, K., Yoshida, Y., Okumura, T., Sato, T., Yokoi, W., & Sawada, H. (2008).
55–60. http://dx.doi.org/10.1016/j.fm.2012.08.012. Preventive effect of Streptococcus thermophilus YIT 2001 on dextran sulfate
FAO/WHO (2002). Food and Agricultural Organization of the United Nations and sodium-induced colitis in mice. Bioscience, Biotechnology, and Biochemistry, 72
World Health Organization. In Joint FAO/WHO working group report on drafting (10), 2543–2547.
guidelines for the evaluation of probiotics in food. Food and Agricultural Ito, M., Ohishi, K., Yoshida, Y., Yokoi, W., & Sawada, H. (2003). Antioxidative effects
Organization of the United Nations <ftp://ftp.fao.org/es/esn/food/wgreport2.pdf>. of lactic acid bacteria on the colonic mucosa of iron-overloaded mice. Journal of
Foligné, B., Daniel, C., & Pot, B. (2013). Probiotics from research to market: The Agricultural and Food Chemistry, 51(15), 4456–4460. http://dx.doi.org/10.1021/
possibilities, risks and challenges. Current Opinion in Microbiology, 16(3), jf0261957.
284–292. http://dx.doi.org/10.1016/j.mib.2013.06.008. Ito, M., Oishi, K., Yoshida, Y., Okumura, T., Sato, T., Naito, E., ... Sawado, H. (2015).
Fontaine, L., & Hols, P. (2008). The inhibitory spectrum of thermophilin 9 from Effects of lactic acid bacteria on low-density lipoprotein susceptibility to
Streptococcus thermophilus LMD-9 depends on the production of multiple oxidation and aortic fatty lesion formation in hyperlipidemic hamsters.
peptides and the activity of BlpG(St), a thiol-disulfide oxidase. Applied and Beneficial Microbes, 6(3), 287–293. http://dx.doi.org/10.3920/BM2014.0040.
Environmental Microbiology, 74(4), 1102–1110. http://dx.doi.org/10.1128/ Ivanova, I., Miteva, V., Stefanova, T., Pantev, A., Budakov, I., Danova, S., ... Boyaval, P.
AEM.02030-07. (1998). Characterization of a bacteriocin produced by Streptococcus
Fox, P. F., McSweeney, P. H. L., Cogan, T. M., & Guinee, T. P. (2004). Cheese: Chemistry, thermophilus 81. International Journal of Food Microbiology, 42(3), 147–158.
physics and microbiology (3rd ed.). London: Academic Press. Retrieved from Iyer, R., Tomar, S. K., Kapila, S., Mani, J., & Singh, R. (2010). Probiotic properties of
<http://store.elsevier.com/Cheese-Chemistry-Physics-and-Microbiology/isbn- folate producing Streptococcus thermophilus strains. Food Research International,
9780122636530/>. 43(1), 103–110. http://dx.doi.org/10.1016/j.foodres.2009.09.011.
Fujii, A., & Cook, E. S. (1973). Probiotics. Antistaphylococcal and antifibrinolytic Jameh, N., Galia, W., Awussi, A. A., Roux, E., Genay, M., Perrin, C., ... Dary-Mourot, A.
activities of omega-guanidino acids and omega-guanidinoacyl-L-histidines. (2016). Characterization of a new peptide transport system in Streptococcus
Journal of Medicinal Chemistry, 16(12), 1409–1411. thermophilus. Food Research International, 86, 34–45. http://dx.doi.org/10.1016/
Fuller, R. (1992). In R. Fuller (Ed.), History and development of probiotics. London, UK: j.foodres.2016.04.039.
Chapman & Hall. Junjua, M., Kechaou, N., Chain, F., Awussi, A. A., Roussel, Y., Perrin, C., ... Dary-
Gao, X.-Y., Zhi, X.-Y., Li, H.-W., Klenk, H.-P., & Li, W.-J. (2014). Comparative genomics Mourot, A. (2016). A large scale in vitro screening of Streptococcus thermophilus
of the bacterial genus streptococcus illuminates evolutionary implications of strains revealed strains with a high anti-inflammatory potential. LWT - Food
species groups. PLoS ONE, 9(6), e101229. http://dx.doi.org/10.1371/journal. Science and Technology, 70, 78–87. http://dx.doi.org/10.1016/j.lwt.2016.02.006.
pone.0101229. Kabuki, T., Uenishi, H., Watanabe, M., Seto, Y., & Nakajima, H. (2007).
García-Albiach, R., Pozuelo de Felipe, M. J., José, M., de Felipe, P., Angulo, S., Characterization of a bacteriocin, Thermophilin 1277, produced by
Morosini, M.-I., ... Del Campo, R. (2008). Molecular analysis of yogurt containing Streptococcus thermophilus SBT1277. Journal of Applied Microbiology, 102(4),
Lactobacillus delbrueckii subsp. bulgaricus and Streptococcus thermophilus in 971–980. http://dx.doi.org/10.1111/j.1365-2672.2006.03159.x.
human intestinal microbiota. The American Journal of Clinical Nutrition, 87(1), Kebouchi, M., Galia, W., Genay, M., Soligot, C., Lecomte, X., Awussi, A. A., ... Le Roux,
91–96. Y. (2016). Implication of sortase-dependent proteins of Streptococcus
García-Hernández, J., Moreno, Y., Chuan, C., & Hernández, M. (2012). In vivo study of thermophilus in adhesion to human intestinal epithelial cell lines and bile salt
the survival of Lactobacillus delbrueckii subsp. bulgaricus CECT 4005T and tolerance. Applied Microbiology and Biotechnology, 100(8), 3667–3679. http://dx.
Streptococcus thermophilus CECT 801 by DVC-FISH after consumption of doi.org/10.1007/s00253-016-7322-1.
fermented milk. Journal of Food Science, 77(10), M593–597. http://dx.doi.org/ Kekkonen, R.-A., Kajasto, E., Miettinen, M., Veckman, V., Korpela, R., & Julkunen, I.
10.1111/j.1750-3841.2012.02918.x. (2008). Probiotic Leuconostoc mesenteroides ssp. cremoris and Streptococcus
Gilbreth, S. E., & Somkuti, G. A. (2005). Thermophilin 110: A bacteriocin of thermophilus induce IL-12 and IFN-gamma production. World Journal of
Streptococcus thermophilus ST110. Current Microbiology, 51(3), 175–182. http:// Gastroenterology, 14(8), 1192–1203.
dx.doi.org/10.1007/s00284-005-4540-7. Kolb, H. (1955). Die Behandlung akuter Infekte unter dem Gesichtswinkel der
Giovannini, M., Agostoni, C., Riva, E., Salvini, F., Ruscitto, A., Zuccotti, G. V., ... Felicita Prophylaxe chronischer Leiden. Ü ber die Behandlung mit physiologischen
Study Group (2007). A randomized prospective double blind controlled trial on bakterien. Microecology Therapy, 1, 15–19.
effects of long-term consumption of fermented milk containing Lactobacillus Kollath, W. (1953). [Nutrition and the tooth system; general review with special
casei in pre-school children with allergic asthma and/or rhinitis. Pediatric reference to vitamins]. Deutsche Zahnärztliche Zeitschrift, 8(11). Suppl 7-16.
Research, 62(2), 215–220. http://dx.doi.org/10.1203/PDR.0b013e3180a76d94. Kolling, G. L., Wu, M., Warren, C. A., Durmaz, E., Klaenhammer, T. R., Timko, M. P., &
González-Márquez, H., Perrin, C., Bracquart, P., Guimont, C., & Linden, G. (1997). A Guerrant, R. L. (2012). Lactic acid production by Streptococcus thermophilus
16 kDa protein family overexpressed by Streptococcus thermophilus PB18 in acid alters Clostridium difficile infection and in vitro Toxin A production. Gut
environments. Microbiology (Reading, England), 143(Pt 5), 1587–1594. Microbes, 3(6), 523–529. http://dx.doi.org/10.4161/gmic.21757.
Guandalini, S., Magazzù, G., Chiaro, A., La Balestra, V., Di Nardo, G., Gopalan, S., ... Korpela, K., Salonen, A., Virta, L. J., Kumpu, M., Kekkonen, R. A., & de Vos, W. M.
Setty, M. (2010). VSL#3 improves symptoms in children with irritable bowel (2016). Lactobacillus rhamnosus GG intake modifies preschool children’s
syndrome: A multicenter, randomized, placebo-controlled, double-blind, intestinal microbiota, alleviates penicillin-associated changes, and reduces
crossover study. Journal of Pediatric Gastroenterology and Nutrition, 51(1), antibiotic use. PLoS ONE, 11(4), e0154012. http://dx.doi.org/10.1371/journal.
24–30. http://dx.doi.org/10.1097/MPG.0b013e3181ca4d95. pone.0154012.
Guillemard, E., Tondu, F., Lacoin, F., & Schrezenmeir, J. (2010). Consumption of a Koyama, T., Kirjavainen, P. V., Fisher, C., Anukam, K., Summers, K., Hekmat, S., &
fermented dairy product containing the probiotic Lactobacillus casei DN- Reid, G. (2010). Development and pilot evaluation of a novel probiotic mixture
114001 reduces the duration of respiratory infections in the elderly in a for the management of seasonal allergic rhinitis. Canadian Journal of
randomised controlled trial. The British Journal of Nutrition, 103(1), 58–68. Microbiology, 56(9), 730–738. http://dx.doi.org/10.1139/w10-061.
http://dx.doi.org/10.1017/S0007114509991395. Latvala, S., Miettinen, M., Kekkonen, R. A., Korpela, R., & Julkunen, I. (2011).
Hasslöf, P., West, C. E., Videhult, F. K., Brandelius, C., & Stecksén-Blicks, C. (2013). Lactobacillus rhamnosus GG and Streptococcus thermophilus induce suppressor
Early intervention with probiotic Lactobacillus paracasei F19 has no long-term of cytokine signalling 3 (SOCS3) gene expression directly and indirectly via
effect on caries experience. Caries Research, 47(6), 559–565. http://dx.doi.org/ interleukin-10 in human primary macrophages. Clinical and Experimental
10.1159/000350524. Immunology, 165(1), 94–103. http://dx.doi.org/10.1111/j.1365-
Hauser, G., Salkic, N., Vukelic, K., JajacKnez, A., & Stimac, D. (2015). Probiotics for 2249.2011.04408.x.
standard triple helicobacter pylori eradication. Medicine, 94(17). http://dx.doi. Latvala, S., Pietila, T.-E., Veckman, V., Kekkonen, R.-A., Tynkkynen, S., Korpela, R., &
org/10.1097/MD.0000000000000685. Julkunen, I. (2008). Potentially probiotic bacteria induce efficient maturation
Havenaar, R., & Huis in’t Veld, J. H. J. (1992). Probiotics a general view (Elsevier but differential cytokine production in human monocyte-derived dendritic
Applied Science, Vol. 1). London: Wood BJB. cells. World Journal of Gastroenterology, 14(36), 5570–5583–5582.
Helinck, S., Le Bars, D., Moreau, D., & Yvon, M. (2004). Ability of thermophilic lactic Leyer, G. J., Li, S., Mubasher, M. E., Reifer, C., & Ouwehand, A. C. (2009). Probiotic
acid bacteria to produce aroma compounds from amino acids. Applied and effects on cold and influenza-like symptom incidence and duration in children.
Environmental Microbiology, 70(7), 3855–3861. http://dx.doi.org/10.1128/ Pediatrics, 124(2), e172–179. http://dx.doi.org/10.1542/peds.2008-2666.
AEM.70.7.3855-3861.2004. Lick, S., Drescher, K., & Heller, K. J. (2001). Survival of Lactobacillus delbrueckii
Hill, C., Guarner, F., Reid, G., Gibson, G. R., Merenstein, D. J., Pot, B., ... Sanders, M. E. subsp. bulgaricus and Streptococcus thermophilus in the Terminal Ileum of
(2014). Expert consensus document. The international scientific association for Fistulated Gottingen Minipigs. Applied and Environmental Microbiology, 67(9),
probiotics and prebiotics consensus statement on the scope and appropriate use 4137–4143. http://dx.doi.org/10.1128/AEM.67.9.4137-4143.2001.
of the term probiotic. Nature Reviews. Gastroenterology & Hepatology, 11(8), Lilly, D. M., & Stillwell, R. H. (1965). Probiotics: Growth-promoting factors produced
506–514. http://dx.doi.org/10.1038/nrgastro.2014.66. by microorganisms. Science (New York, NY), 147(3659), 747–748.
Hols, P., Hancy, F., Fontaine, L., Grossiord, B., Prozzi, D., Leblond-Bourget, N., ... Liu, M., Siezen, R. J., & Nauta, A. (2009). In silico prediction of horizontal gene
Kleerebezem, M. (2005). New insights in the molecular biology and physiology transfer events in Lactobacillus bulgaricus and Streptococcus thermophilus
of Streptococcus thermophilus revealed by comparative genomics. FEMS reveals protocooperation in yogurt manufacturing. Applied and Environmental
Microbiology Reviews, 29(3), 435–463. http://dx.doi.org/10.1016/j. Microbiology, 75(12), 4120–4129. http://dx.doi.org/10.1128/AEM.02898-08.
femsre.2005.04.008. Makarova, K., Slesarev, A., Wolf, Y., Sorokin, A., Mirkin, B., Koonin, E., ... Mills, D.
Hurduc, V., Plesca, D., Dragomir, D., Sajin, M., & Vandenplas, Y. (2009). A (2006). Comparative genomics of the lactic acid bacteria. Proceedings of the
randomized, open trial evaluating the effect of Saccharomyces boulardii on National Academy of Sciences of the United States of America, 103(42),
the eradication rate of Helicobacter pylori infection in children. Acta Paediatrica 15611–15616. http://dx.doi.org/10.1073/pnas.0607117103.
88 O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89

Makino, S., Ikegami, S., Kume, A., Horiuchi, H., Sasaki, H., & Orii, N. (2010). Reducing tract of healthy and hypochlorhydric elderly subjects. The American Journal of
the risk of infection in the elderly by dietary intake of yoghurt fermented with Clinical Nutrition, 61(2), 353–359.
Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1. The British Journal of Perez, P. F., de Antoni, G. L., & Añon, M. C. (1991). Formate production by
Nutrition, 104(7), 998–1006. http://dx.doi.org/10.1017/S000711451000173X. Streptococcus thermophilus cultures. Journal of Dairy Science, 74(9), 2850–2854.
Marciset, O., Jeronimus-Stratingh, M. C., Mollet, B., & Poolman, B. (1997). http://dx.doi.org/10.3168/jds.S0022-0302(91)78465-8.
Thermophilin 13, a nontypical antilisterial poration complex bacteriocin, that Pitino, I., Randazzo, C. L., Cross, K. L., Parker, M. L., Bisignano, C., Wickham, M. S. J., ...
functions without a receptor. The Journal of Biological Chemistry, 272(22), Caggia, C. (2012). Survival of Lactobacillus rhamnosus strains inoculated in
14277–14284. cheese matrix during simulated human digestion. Food Microbiology, 31(1),
Marteau, P., Minekus, M., Havenaar, R., & Huis in’t Veld, J. H. (1997). Survival of 57–63. http://dx.doi.org/10.1016/j.fm.2012.02.013.
lactic acid bacteria in a dynamic model of the stomach and small intestine: Pochart, P., Dewit, O., Desjeux, J.-F., & Bourlioux, P. (1989). Viable starter culture,
Validation and the effects of bile. Journal of Dairy Science, 80(6), 1031–1037. beta-galactosidase activity, and lactose in duodenum after yogurt ingestion in
http://dx.doi.org/10.3168/jds.S0022-0302(97)76027-2. lactase-deficient humans. American Journal Clinical Nutrition, 49, 828–831.
Marzorati, M., Vanhoecke, B., De Ryck, T., Sadaghian Sadabad, M., Pinheiro, I., Possemiers, S., Marzorati, M., Verstraete, W., & Van de Wiele, T. (2010). Bacteria and
Possemiers, S., ... Van de Wiele, T. (2014). The HMITM module: a new tool to chocolate: A successful combination for probiotic delivery. International
study the Host-Microbiota Interaction in the human gastrointestinal tract Journal of Food Microbiology, 141(1–2), 97–103. http://dx.doi.org/10.1016/j.
in vitro. BMC Microbiology, 14, 133. http://dx.doi.org/10.1186/1471-2180-14- ijfoodmicro.2010.03.008.
133. Preidis, G. A., Hill, C., Guerrant, R. L., Ramakrishna, B. S., Tannock, G. W., & Versalovic,
Mater, D. D., Bretigny, L., Firmesse, O., Flores, M.-J., Mogenet, A., Bresson, J.-L., & J. (2011). Probiotics, enteric and diarrheal diseases, and global health.
Corthier, G. (2005). Streptococcus thermophilus and Lactobacillus delbrueckii Gastroenterology, 140(1), 8–14. http://dx.doi.org/10.1053/j.gastro.2010.11.010.
subsp. bulgaricus survive gastrointestinal transit of healthy volunteers Rasmussen, T. B., Danielsen, M., Valina, O., Garrigues, C., Johansen, E., & Pedersen, M.
consuming yogurt. FEMS Microbiology Letters, 250(2), 185–187. http://dx.doi. B. (2008). Streptococcus thermophilus core genome: Comparative genome
org/10.1016/j.femsle.2005.07.006. hybridization study of 47 strains. Applied and Environmental Microbiology, 74
Mater, D. D., Drouault-Holowacz, S., Oozeer, R., Langella, P., Anba, J., & Corthier, G. (15), 4703–4710. http://dx.doi.org/10.1128/AEM.00132-08.
(2006). Β-Galactosidase production by Streptococcus thermophilus is higher in Resta-Lenert, S., & Barrett, K. E. (2003). Live probiotics protect intestinal epithelial
the small intestine than in the caecum of human-microbiota-associated mice cells from the effects of infection with Enteroinvasive Escherichia coli (EIEC).
after lactose supplementation. British Journal of Nutrition, 96(01), 177–181. Gut, 52(7), 988–997.
Mathot, A. G., Beliard, E., & Thuault, D. (2003). Streptococcus thermophilus 580 Rodríguez, C., Medici, M., Mozzi, F., & Font de Valdez, G. (2010). Therapeutic effect of
produces a bacteriocin potentially suitable for inhibition of Clostridium Streptococcus thermophilus CRL 1190-fermented milk on chronic gastritis. World
tyrobutyricum in hard cheese. Journal of Dairy Science, 86(10), 3068–3074. Journal of Gastroenterology, 16(13), 1622–1630.
http://dx.doi.org/10.3168/jds.S0022-0302(03)73906-X. Rodríguez, C., Medici, M., Rodríguez, A. V., Mozzi, F., & Font de Valdez, G. (2009).
Metchnikoff, E. (1907). The prolongation of life; optimistic studies. London: Prevention of chronic gastritis by fermented milks made with
Heinemann. exopolysaccharide-producing Streptococcus thermophilus strains. Journal of
Minekus, M., Marteau, P., Havenaar, R., & Huis in’t Veld, J. H. (1995). A Dairy Science, 92(6), 2423–2434. http://dx.doi.org/10.3168/jds.2008-1724.
multicompartmental dynamic computer-controlled model simulating the Rossi, F., Marzotto, M., Cremonese, S., Rizzotti, L., & Torriani, S. (2013). Diversity of
stomach and small intestine. ATLA, 23, 197–209. Streptococcus thermophilus in bacteriocin production; inhibitory spectrum and
Minelli, E. B., & Benini, A. (2008). Relationship between number of bacteria and their occurrence of thermophilin genes. Food Microbiology, 35(1), 27–33. http://dx.
probiotic effects. Microbial Ecology in Health and Disease, 20(4), 180–183. http:// doi.org/10.1016/j.fm.2013.02.006.
dx.doi.org/10.1080/08910600802408095. Rul, F., Ben-Yahia, L., Chegdani, F., Wrzosek, L., Thomas, S., Noordine, M.-L., ...
Miquel, S., Beaumont, M., Martín, R., Langella, P., Braesco, V., & Thomas, M. (2015). A Thomas, M. (2011). Impact of the Metabolic Activity of Streptococcus
proposed framework for an appropriate evaluation scheme for microorganisms thermophilus on the Colon Epithelium of Gnotobiotic Rats. Journal of Biological
as novel foods with a health claim in Europe. Microbial Cell Factories, 14, 48. Chemistry, 286(12), 10288–10296. http://dx.doi.org/10.1074/jbc.M110.168666.
http://dx.doi.org/10.1186/s12934-015-0229-1. Saavedra, J. M., Abi-Hanna, A., Moore, N., & Yolken, R. H. (2004). Long-term
Molly, K., Vande Woestyne, M., & Verstraete, W. (1993). Development of a 5-step consumption of infant formulas containing live probiotic bacteria: tolerance
multi-chamber reactor as a simulation of the human intestinal microbial and safety. The American Journal of Clinical Nutrition, 79(2), 261–267.
ecosystem. Applied Microbiology and Biotechnology, 39(2), 254–258. Saavedra, J. M., Bauman, N. A., Oung, I., Perman, J. A., & Yolken, R. H. (1994). Feeding
Mozzi, F., Gerbino, E., Font de Valdez, G., & Torino, M. I. (2009). Functionality of of Bifidobacterium bifidum and Streptococcus thermophilus to infants in hospital
exopolysaccharides produced by lactic acid bacteria in an in vitro gastric for prevention of diarrhoea and shedding of rotavirus. Lancet (London, England),
system. Journal of Applied Microbiology, 107(1), 56–64. http://dx.doi.org/ 344(8929), 1046–1049.
10.1111/j.1365-2672.2009.04182.x. Salminen, S. (1996). Uniqueness of probiotic strains. IDF Nutrition Newsletter, 5,
Nagpal, R., Kumar, A., Kumar, M., Behare, P. V., Jain, S., & Yadav, H. (2012). Probiotics, 16–18.
their health benefits and applications for developing healthier foods: a review. Salminen, S., Ouwehand, A., Benno, Y., & Lee, Y. K. (1999). Probiotics: how should
FEMS Microbiology Letters, 334(1), 1–15. http://dx.doi.org/10.1111/j.1574- they be defined? Trends in Food Science & Technology, 10(3), 107–110. http://dx.
6968.2012.02593.x. doi.org/10.1016/S0924-2244(99)00027-8.
Näse, L., Hatakka, K., Savilahti, E., Saxelin, M., Pönkä, A., Poussa, T., ... Meurman, J. H. Schaafsma, G. (1996). State-of-the-art concerning probiotic strains in milk products.
(2001). Effect of long-term consumption of a probiotic bacterium, Lactobacillus IDF Nutrition Newsletter, 5, 23–25.
rhamnosus GG, in milk on dental caries and caries risk in children. Caries Schrezenmeir, J., & de Vrese, M. (2001). Probiotics, prebiotics, and synbiotics–
Research, 35(6), 412–420. approaching a definition. The American Journal of Clinical Nutrition, 73(2 Suppl),
O’Callaghan, A., & van Sinderen, D. (2016). Bifidobacteria and their role as members 361S–364S.
of the human gut microbiota. Frontiers in Microbiology, 7. http://dx.doi.org/ Senok, A. C., Ismaeel, A. Y., & Botta, G. A. (2005). Probiotics: Facts and myths. Clinical
10.3389/fmicb.2016.00925. Microbiology and Infection, 11(12), 958–966. http://dx.doi.org/10.1111/j.1469-
Ogita, T., Bergamo, P., Maurano, F., D’Arienzo, R., Mazzarella, G., Bozzella, G., ... Rossi, 0691.2005.01228.x.
M. (2015). Modulatory activity of Lactobacillus rhamnosus OLL2838 in a mouse Shah, N. P. (2007). Functional cultures and health benefits. International Dairy
model of intestinal immunopathology. Immunobiology, 220(6), 701–710. http:// Journal, 17(11), 1262–1277. http://dx.doi.org/10.1016/j.idairyj.2007.01.014.
dx.doi.org/10.1016/j.imbio.2015.01.004. Sieuwerts, S., de Bok, F. A. M., Hugenholtz, J., & van Hylckama Vlieg, J. E. T. (2008).
Ogita, T., Nakashima, M., Morita, H., Saito, Y., Suzuki, T., & Tanabe, S. (2011). Unraveling microbial interactions in food fermentations: From classical to
Streptococcus thermophilus ST28 ameliorates colitis in mice partially by genomics approaches. Applied and Environmental Microbiology, 74(16),
suppression of inflammatory Th17 cells. Journal of Biomedicine & 4997–5007. http://dx.doi.org/10.1128/AEM.00113-08.
Biotechnology, 2011, 378417. http://dx.doi.org/10.1155/2011/378417. Sokol, H., Pigneur, B., Watterlot, L., Lakhdari, O., Bermúdez-Humarán, L. G.,
Ogita, T., Tanii, Y., Morita, H., Suzuki, T., & Tanabe, S. (2011). Suppression of Th17 Gratadoux, J.-J., ... Langella, P. (2008). Faecalibacterium prausnitzii is an anti-
response by Streptococcus thermophilus ST28 through induction of IFN-c. inflammatory commensal bacterium identified by gut microbiota analysis of
International Journal of Molecular Medicine, 28(5), 817–822. http://dx.doi.org/ Crohn disease patients. Proceedings of the National Academy of Sciences of the
10.3892/ijmm.2011.755. United States of America, 105(43), 16731–16736. http://dx.doi.org/10.1073/
Palma, M. L., Zamith-Miranda, D., Martins, F. S., Bozza, F. A., Nimrichter, L., Montero- pnas.0804812105.
Lomeli, M., ... Douradinha, B. (2015). Probiotic Saccharomyces cerevisiae strains Sperti, G. S. (1971). Probiotics. West Point, Connecticut: AVI Publishing Co., Inc.
as biotherapeutic tools: Is there room for improvement? Applied Microbiology Stensson, M., Koch, G., Coric, S., Abrahamsson, T. R., Jenmalm, M. C., Birkhed, D., &
and Biotechnology, 99(16), 6563–6570. http://dx.doi.org/10.1007/s00253-015- Wendt, L.-K. (2014). Oral administration of Lactobacillus reuteri during
6776-x. the first year of life reduces caries prevalence in the primary dentition at
Parker, R. B. (1974). Probiotics, the other half of the story. Animal Nutrition and 9 years of age. Caries Research, 48(2), 111–117. http://dx.doi.org/10.1159/
Health, 29, 4–8. 000354412.
Patro-Golab, B., Shamir, R., & Szajewska, H. (2015). Yogurt for treating antibiotic- Sun, Z., Chen, X., Wang, J., Zhao, W., Shao, Y., Wu, L., ... Chen, W. (2011). Complete
associated diarrhea: Systematic review and meta-analysis. Nutrition (Burbank, genome sequence of Streptococcus thermophilus strain ND03. Journal of
Los Angeles County, Calif.), 31(6), 796–800. http://dx.doi.org/10.1016/j. Bacteriology, 193(3), 793–794. http://dx.doi.org/10.1128/JB.01374-10.
nut.2014.11.013. Teanpaisan, R., & Piwat, S. (2014). Lactobacillus paracasei SD1, a novel probiotic,
Pedrosa, M. C., Golner, B. B., Goldin, B. R., Barakat, S., Dallal, G. E., & Russell, R. M. reduces mutans streptococci in human volunteers: A randomized placebo-
(1995). Survival of yogurt-containing organisms and Lactobacillus gasseri controlled trial. Clinical Oral Investigations, 18(3), 857–862. http://dx.doi.org/
(ADH) and their effect on bacterial enzyme activity in the gastrointestinal 10.1007/s00784-013-1057-5.
O. Uriot et al. / Journal of Functional Foods 37 (2017) 74–89 89

Terahara, M., Kurama, S., & Takemoto, N. (2001). Prevention by lactic acid bacteria Vieira, A. T., Galvão, I., Amaral, F. A., Teixeira, M. M., Nicoli, J. R., & Martins, F. S.
of the oxidation of human LDL. Bioscience, Biotechnology, and Biochemistry, 65(8), (2015). Oral treatment with Bifidobacterium longum 51 A reduced
1864–1868. inflammation in a murine experimental model of gout. Beneficial Microbes, 6
Thibessard, A., Borges, F., Fernandez, A., Gintz, B., Decaris, B., & Leblond-Bourget, N. (6), 799–806. http://dx.doi.org/10.3920/BM2015.0015.
(2004). Identification of Streptococcus thermophilus CNRZ368 genes involved in Villani, F., Pepe, O., Mauriello, G., Salzano, G., Moschetti, G., & Coppola, S. (1995).
defense against superoxide stress. Applied and Environmental Microbiology, 70 Antilisterial activity of thermophilin 347, a bacteriocin produced by
(4), 2220–2229. Streptococcus thermophilus. International Journal of Food Microbiology, 25(2),
Thomas, M., Wrzosek, L., Ben-Yahia, L., Noordine, M.-L., Gitton, C., Chevret, D., ... Rul, 179–190.
F. (2011). Carbohydrate metabolism is essential for the colonization of Vinderola, C. G., & Reinheimer, J. A. (2003). Lactic acid starter and probiotic bacteria:
Streptococcus thermophilus in the digestive tract of gnotobiotic rats. PLoS ONE, A comparative ‘‘in vitro” study of probiotic characteristics and biological barrier
6(12), e28789. http://dx.doi.org/10.1371/journal.pone.0028789. resistance. Food Research International, 36(9–10), 895–904. http://dx.doi.org/
Tian, F., Chi, F., Wang, G., Liu, X., Zhang, Q., Chen, Y., ... Chen, W. (2015). Lactobacillus 10.1016/S0963-9969(03)00098-X.
rhamnosus CCFM1107 treatment ameliorates alcohol-induced liver injury in a Wang, Y., & Huang, Y. (2014). Effect of Lactobacillus acidophilus and
mouse model of chronic alcohol feeding. Journal of Microbiology (Seoul, Korea), Bifidobacterium bifidum supplementation to standard triple therapy on
53(12), 856–863. http://dx.doi.org/10.1007/s12275-015-5239-5. Helicobacter pylori eradication and dynamic changes in intestinal flora. World
Tian, H., Tan, J., Zhang, L., Gu, X., Xu, W., Guo, X., & Luo, Y. (2012). Increase of stress Journal of Microbiology & Biotechnology, 30(3), 847–853. http://dx.doi.org/
resistance in Lactococcus lactis via a novel food-grade vector expressing a shsp 10.1007/s11274-013-1490-2.
gene from Streptococcus thermophilus. Brazilian Journal of Microbiology: Wang, B., Xu, H., Xu, F., Zeng, Z., & Wei, H. (2016). Efficacy of oral Bifidobacterium
[Publication of the Brazilian Society for Microbiology], 43(3), 1157–1164. http:// bifidum ATCC 29521 on microflora and antioxidant in mice. Canadian Journal of
dx.doi.org/10.1590/S1517-838220120003000043. Microbiology, 62(3), 249–262. http://dx.doi.org/10.1139/cjm-2015-0685.
Trabelsi, I., Ktari, N., Ben Slima, S., Hamden, K., & Ben Salah, R. (2016). Effect of a Ward, D. J., & Somkuti, G. A. (1995). Characterization of a bacteriocin produced by
probiotic Lactobacillus plantarum TN8 strain on trinitrobenzene sulphonic acid- Streptococcus thermophilus ST134. Applied Microbiology and Biotechnology, 43(2),
induced colitis in rats. Journal of Animal Physiology and Animal Nutrition. http:// 330–335.
dx.doi.org/10.1111/jpn.12536. West, C. E., Hammarström, M.-L., & Hernell, O. (2009). Probiotics during weaning
Trip, H., Mulder, N. L., & Lolkema, J. S. (2012). Improved acid stress survival of reduce the incidence of eczema. Pediatric Allergy and Immunology: Official
Lactococcus lactis expressing the histidine decarboxylation pathway of Publication of the European Society of Pediatric Allergy and Immunology, 20(5),
Streptococcus thermophilus CHCC1524. The Journal of Biological Chemistry, 287 430–437. http://dx.doi.org/10.1111/j.1399-3038.2009.00745.x.
(14), 11195–11204. http://dx.doi.org/10.1074/jbc.M111.330704. Ziar, H., Gérard, P., & Riazi, A. (2014). Effect of prebiotic carbohydrates on growth,
Uriot, O., Galia, W., Awussi, A. A., Perrin, C., Denis, S., Chalancon, S., ... Roussel, Y. bile survival and cholesterol uptake abilities of dairy-related bacteria. Journal of
(2016). Use of the dynamic gastro-intestinal model TIM to explore the survival the Science of Food and Agriculture, 94(6), 1184–1190. http://dx.doi.org/10.1002/
of the yogurt bacterium Streptococcus thermophilus and the metabolic activities jsfa.6395.
induced in the simulated human gut. Food Microbiology, 53(Pt A), 18–29. http:// Ziarno, M. (2010). Viability and cholesterol uptake by Streptococcus thermophilus
dx.doi.org/10.1016/j.fm.2015.05.007. cultures in artificial GIT fluids. Acta Scientiarum Polonorum Technologia
Vasiljevic, T., & Shah, N. P. (2008). Probiotics—from metchnikoff to bioactives. Alimentaria, 9(1).
International Dairy Journal, 18(7), 714–728. http://dx.doi.org/10.1016/j. Zotta, T., Ricciardi, A., Ciocia, F., Rossano, R., & Parente, E. (2008). Diversity of stress
idairyj.2008.03.004. responses in dairy thermophilic streptococci. International Journal of Food
Venturi, A., Gionchetti, P., Rizzello, F., Johansson, R., Zucconi, E., Brigidi, P., ... Microbiology, 124(1), 34–42. http://dx.doi.org/10.1016/j.
Campieri, M. (1999). Impact on the composition of the faecal flora by a new ijfoodmicro.2008.02.024.
probiotic preparation: preliminary data on maintenance treatment of patients Zotta, T., Ricciardi, A., Rossano, R., & Parente, E. (2008). Urease production by
with ulcerative colitis. Alimentary Pharmacology & Therapeutics, 13(8), Streptococcus thermophilus. Food Microbiology, 25(1), 113–119. http://dx.doi.org/
1103–1108. 10.1016/j.fm.2007.07.001.
Vergin, F. (1954). Antibiotics and probiotics. Hippokrates, 25(4), 116–119.

You might also like