ArticlePDF Available

An Infant Milk Formula Supplemented with Heat-Treated Probiotic Bifidobacterium animalis subsp. lactis CECT 8145, Reduces Fat Deposition in C. elegans and Augments Acetate and Lactate in a Fermented Infant Slurry

Authors:
  • ADM Biopolis, Valencia, Spain

Abstract

Pediatric obesity has a growing health and socio-economical impact due to cardiovascular and metabolic complications in adult life. Some recent studies suggest that live or heat-treated probiotics have beneficial effects in preventing fat deposition and obesity in preclinical and clinical sets. Here, we have explored the effects of heat-treated probiotic Bifidobacterium animalis subsp. lactis CECT 8145 (HT-BPL1), added as a supplement on an infant milk formula (HT-BPL1-IN), on Caenorhabditis elegans fat deposition and short-chain fatty acids (SCFAs) and lactate, using fermented baby fecal slurries. We have found that HT-BPL1-IN significantly reduced fat deposition in C. elegans, at the time it drastically augmented the generation of some SCFAs, particulary acetate and organic acid lactate. Data suggest that heat-treated BPL1 maintains its functional activities when added to an infant powder milk formula.
foods
Article
An Infant Milk Formula Supplemented with
Heat-Treated Probiotic Bifidobacterium animalis
subsp. lactis CECT 8145, Reduces Fat Deposition in
C. elegans and Augments Acetate and Lactate in a
Fermented Infant Slurry
Ángela Silva 1, , Nuria Gonzalez 1, , Ana Terrén2, Antonio García2,3,4,
Juan Francisco Martinez-Blanch 5, Vanessa Illescas 5, Javier Morales 6, Marcos Maroto 2,
Salvador Genovés1, Daniel Ramón1,5, Patricia Martorell 1and Empar Chenoll 1, 5, *
1Health & Wellness-ADM Nutrition-ADM Biopolis, 46980 Paterna, Spain; angela.silva@adm.com (Á.S.);
nuria.gonzalez@adm.com (N.G.); Salvador.genoves@adm.com (S.G.); daniel.ramonvidal@adm.com (D.R.);
patricia.martorell@adm.com (P.M.)
2Instituto Fundación Teófilo Hernando, 28029 Madrid, Spain; ana.terren@ifth.es (A.T.); agg@uam.es (A.G.);
marcos.maroto@ifth.es (M.M.)
3Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, 28029 Madrid, Spain
4Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Facultad de Medicina,
28006 Madrid, Spain
5Health & Wellness-ADM Nutrition-ADM LIfesequencing, 46980 Paterna, Spain;
juan.martinezblanch@adm.com (J.F.M.-B.); Vanessa.illescas@adm.com (V.I.)
6Product Development Department, Alter Farmacia SA, 28880 Madrid, Spain; jmoraleso@grupo-alter.com
*Correspondence: maria.chenoll@adm.com
These two authors have contributed equally to the present work.
Received: 20 April 2020; Accepted: 13 May 2020; Published: 19 May 2020


Abstract:
Pediatric obesity has a growing health and socio-economical impact due to cardiovascular
and metabolic complications in adult life. Some recent studies suggest that live or heat-treated
probiotics have beneficial eects in preventing fat deposition and obesity in preclinical and clinical
sets. Here, we have explored the eects of heat-treated probiotic Bifidobacterium animalis subsp.
lactis CECT 8145 (HT-BPL1), added as a supplement on an infant milk formula (HT-BPL1-IN),
on Caenorhabditis elegans fat deposition and short-chain fatty acids (SCFAs) and lactate, using
fermented baby fecal slurries. We have found that HT-BPL1-IN significantly reduced fat deposition
in C. elegans, at the time it drastically augmented the generation of some SCFAs, particulary acetate
and organic acid lactate. Data suggest that heat-treated BPL1 maintains its functional activities when
added to an infant powder milk formula.
Keywords:
heat-treated probiotic; Bifidobacterium animalis subsp. lactis; fat deposition; acetate; lactate;
C. elegans
1. Introduction
A pool analysis of 128.9 million children, adolescents, and adults reveals the huge health impact
of obesity [
1
]. This is illustrated by several studies. For instance, adults with a history of chilhood
obesity have higher prevalence of metabolic syndrome and cardiovascular risk [
2
,
3
]. Additional
complications of pediatric obesity are poor skeletal maturation [
4
], psychosocial inadaptation [
5
],
or early inflammatory status [6].
Foods 2020,9, 652; doi:10.3390/foods9050652 www.mdpi.com/journal/foods
Foods 2020,9, 652 2 of 13
During the last years, new strategies derived from the gut microbiome analysis are emerging
to understand and combat childhood obesity. Thus, an association of early life gut microbiota
with body mass index has been demonstrated in an observational study in 0–3 years age infants [
7
].
Such association seems to be linked to short-chain fatty acids (SCFA) binding G protein coupled receptor
(GPR41), which modulates host adiposity [
8
]. These findings point to a potential use of prebiotic,
probiotic or symbiotic products to modify in some way the gut microbiota in individuals prone to
obesity and to reduce the impact of this dysbiosis in childhood and later adulthood. Specific probiotic
supplements can impinge on lower incidence of obesity and hypertension [
9
12
]. Other studies have
also shown that specific probiotics impact on weight management in adults [1012]
Recent studies have shown that heat-treated bacteria release metabolites or cell components
that can display functional actions, such as immunomodulation or antagonism of pathogens [
13
15
].
This has led to an increasing interest in using heat-treated probiotics [
13
16
]. In fact, a few clinical
trials have shown heat-treated bacteria functional activities in for instance infantile colic [
17
]) or in
functional bloating [18].
In a set of preclinical studies, strain Bifidobacterium animalis subsp. lactis strain BPL1 was isolated
from infant feces and deposited in Spanish Type Culture Collection (CECT) under accession number
CECT 8145 and, in a massive screening, demonstrated its capacity to reduce fat deposition [
19
].
Initially, the strain was tested for its ability to reduce fat content in Caenorhabditis elegans model [
19
],
and later in two murine models of obesity; one carried out in conventional wistar rats fed on a cafeteria
diet [
20
] and the second based on the use of genetically obese rats Zücker rats [
21
]. In all three
studies, the animals were fed BPL1 from infancy/larvae state to young adulthood, and in all three cases,
an eect of lifetime probiotic consumption on adult fat was observed. In the two murine model studies,
it was also observed that probiotic intake in lean individuals had no eect on their body fat levels
or other measured parameters. Moreover, both in the C. elegans model and in the study in rodents
on a cafeteria diet, promising results were obtained with a thermally treated version of the probiotic
strain, pointing to a non-life-dependent mode of action, as a part of BPL1 eectiveness. However,
while these results showed an eect of BPL1 cells on fat deposition in both animal models and clinical
human trial, we were unable to study the potential involvement of the gut microbiome in the observed
functionality. This potential involvement is of great importance in order to be able to analyze in depth
and systematically the potential mechanisms of action of the probiotic strain (by direct action and by
the modulating eect of the microbiome). Thus, further to these results, in this study we considered to
evaluate whether the eect observed in animals and humans was due to a direct action of the cells,
or if there was an impact on the bacterial microbiome and compounds derived from its metabolism
that could have an eect on its functionality in the preclinical model of C. elegans. Secondly, and in
conjunction with the previous point, we defined as an objective to study whether the addition of the
probiotic to a commercial product (in this case infant milk formula) could have a positive or negative
impact (reinforcing or diminishing functionality respectively) on its eect.
In the present study, a mixed fecal slurry model was used to analyze the impact of HT-BPL1 strain
and infant formula containing the heat-treated probiotic on microbiome, and its potential synergistic
eect with the infant milk formula. Fecal slurry approach enabled us to ex vivo analyze together with
microbiome changes by massive sequencing, SCFAs production and, furthermore, the impact of this
microbiome modulation on C. elegans fat deposition.
2. Materials and Methods
2.1. In Vitro Fecal Fermentations
Bifidobacterium lactis BPL1 cells were grown anaerobically in de Man, Rogosa, and Sharpe medium
(MRS; Oxoid, Basingstoke, UK), supplemented with cysteine (0.05% wt/vol; Sigma, St. Louis, MO,
USA; MRS-C) at 37
C for 18 h. Cells were then harvested by centrifuging and viability evaluated
by plate counting in MRS-C agar (anaerobically, 37
C for 48 h). After that, cells were heat treated
Foods 2020,9, 652 3 of 13
(autoclaved at 121
C for 20 min) and the absence of viability checked by broth culturing in MRS
anaerobically at 37
C for 18 h. Fecal fermentations were carried out by using 3 mL fermentations in
10 mL fermentor (24 multi-well plate;
µ
-24 Bioreactor, Applikon Biotechnology, Delft, The Netherlands).
Fresh fecal samples came from 3 months babies fed with a milk formula free of probiotics. Immediately
after being obtained, stools were vacuum stored in refrigerated conditions until the fermentation
experiments were performed. Fresh feces were weighted and dissolved in pre-reduced Mc Bain
and MacFarlane medium [
22
] in a ratio 1:5 (feces:medium) to hydrate them. Three milliliters of
the homogenized mixture (fecal slurry) were inoculated in each fermentor, and heat-treated BPL1
(HT-BPL1) was added alone (HT-BPL1), or included in the infant formula Innova, from Nutriben,
Alter, Madrid, Spain (HT-BPL1-IN). Medium was previously degassed and headspace was removed
before starting fermentation with nitrogen. All the protocol was performed in an anaerobic cabinet.
All fermentors were maintained under a head space of oxygen-free nitrogen and were continuously
stirred. Anaerobiosis, temperature (37
C) and pH (pH 5.5) were maintained constant along the
fermentation period that was run for 24 h. Slurry samples were anaerobically taken and aliquoted for
further processing and analysis.
2.2. Body Fat Monitoring in C. elegans
Fresh samples were analysed in C. elegans model. The eects of fecal slurry samples subjected
to dierent treatments, on body fat content in C. elegans, were analysed as previously described [
19
].
Briefly, concentrated C. elegans cultures (50
µ
L, OD =30) were added on top of a NGM surface
previously seeded with Escherichia coli OP50, to ensure adequate nutrition conditions. Positive
controls were NGM plates with anti-obesity drug Orlistat (6 µg/mL).
Lipid content in worms was determined by adding 0.05
µ
g/mL of Nile red to NGM agar plates
containing the dierent fecal treatments. Worms were incubated for 3 days at 20
C, until they reach
the adult stage. Then, nematode samples placed in M9 buer were assayed for their fluorescence in
a FP-6200 system (JASCO Analytical Instruments, Easton, MD, USA). Excitation wavelength was at
480 nm and emission was at 571 nm. A total of 120 worms per condition were analyzed in each of
2 dierent experiments.
2.3. Organic Acids Analysis
To analyze short chain fatty acids (SFCAs) acetate, propionate, butyrate and succinate, and organic
acid lactate at the end of the experiment, a volume of 500
µ
L of fermentation was centrifuged at
14,000 rpm for 60 min, filtered through a Millipore 0.45
µ
m pore-size filter (Billerica, MA, USA),
and diluted
1
/
2
in MilliQ quality water. The quantification was conducted on a HPLC Acquity equipped
with an Aminex HPX-87H 300
×
7.8 mm column (BioRad) under conditions defined by manufacturer.
Detection was achieved by a refractive index detector. The eluent was H
2
SO
4
5 mM with an isocratic
flow rate of 0.6 mL/min. No internal standard was included. Quantification was performed by the use
of standard curves with correlation coecient R2>0.999.
2.4. Microbiome Analysis by Deep Sequencing
Slurry samples were immediately frozen and stored at
20
C until their processing. DNA from
samples was isolated following Yuang and co-workers [
23
] with minor modifications to avoid bias
in DNA purification toward misrepresentation of gram positive bacteria, with the aid of MagnaPure
Compact System (Roche Life Science). For massive sequencing, the hypervariable region V3-V4 of
the bacterial 16s rRNA gene was amplified using key-tagged eubacterial primers [24] and sequenced
with a MiSeq Illumina Platform, following the Illumina recommendations for Library preparation
and sequencing.
The resulting sequences were split, taking into account the barcode introduced during the PCR
reaction. Quality control of the sequences was performed in dierent steps. Firstly, quality filtering
(with a minimum threshold of Q20) was performed using fastx tool kit version 0.013, then primer
Foods 2020,9, 652 4 of 13
(16s rRNA primers) trimming and length selection (reads over 300 nts) was done with cutadapt version
1.2. (https://cutadapt.readthedocs.io/en/stable) [
25
]. FASTQ files were converted to FASTA files and
UCHIME program version 7.0.1001 [
26
] was used in order to remove chimeras that may arise during
the amplification and sequencing step. Then clean FASTA files were BLAST against NCBI 16s rRNA
database using blastn version 2.2.29+. The resulting XML files were processed using a python script
developed by Lifesequencing S.L. (Paterna, Valencia, Spain) in order to annotate each sequence at
dierent phylogenetic levels (phylum, family, genus and species). Alpha diversity was conducted
using specaccum program in the vegan package, as implemented for R version 3.2.3. Presence results
are expressed as the percentage of each group sequences.
2.5. Statistical Analysis
Results are given as the mean
±
standard deviation. Dierences between the groups were
analyzed using Tukey’s multiple comparison tests. Data on fat deposition in C. elegans were analyzed
by Student’s ttest. Statistical analyses were performed with GraphPad Prism 8.4.1 software (San Diego,
CA, USA), setting the level of statistical significance at 5%.
3. Results
3.1. Eects of a Milk Powder Supplemented with Heat-Treated BPL1, on C. elegans Fat Deposition
First we explored whether an infant milk powder added alone (MP) or MP supplemented with
HT-BPL1 (HT-BPL1-IN, INNOVA), exerted a functional eect on C. elegans fat content. Worms were
incubated by 3 days with milk powder at increasing concentrations. Figure 1shows that fat body content
(monitored as relative fluorescence of Nile Red, ordinate) was unchanged at the four concentrations of
control MP. However, in the case of worms exposed to HT-BPL1-IN, there was a significant 12.5
±
1.7%
reduction (p<0.001) at the concentration of 1.80 mg/mL and 9.0
±
1.4% reduction at the concentration
of 3.60 mg/mL (p<0.001). This preliminary result prompted another set of experiments through the
incubation of fermented baby slurries in the presence of HT-BPL1-IN.
Foods 2020, 9, x FOR PEER REVIEW 10 of 13
version 1.2. (https://cutadapt.readthedocs.io/en/stable) [25]. FASTQ files were converted to FASTA
files and UCHIME program version 7.0.1001 [26] was used in order to remove chimeras that may
arise during the amplification and sequencing step. Then clean FASTA files were BLAST against
NCBI 16s rRNA database using blastn version 2.2.29+. The resulting XML files were processed using
a python script developed by Lifesequencing S.L. (Paterna, Valencia, Spain) in order to annotate each
sequence at different phylogenetic levels (phylum, family, genus and species). Alpha diversity was
conducted using specaccum program in the vegan package, as implemented for R version 3.2.3.
Presence results are expressed as the percentage of each group sequences.
2.5. Statistical Analysis
Results are given as the mean ± standard deviation. Differences between the groups were
analyzed using Tukey’s multiple comparison tests. Data on fat deposition in C. elegans were analyzed
by Student’s t test. Statistical analyses were performed with GraphPad Prism 8.4.1 software (San
Diego, CA, USA), setting the level of statistical significance at 5%.
3. Results
3.1. Effects of a Milk Powder Supplemented with Heat-Treated BPL1, on C. elegans Fat Deposition
First we explored whether an infant milk powder added alone (MP) or MP supplemented with
HT-BPL1 (HT-BPL1-IN, INNOVA), exerted a functional effect on C. elegans fat content. Worms were
incubated by 3 days with milk powder at increasing concentrations. Figure 1 shows that fat body
content (monitored as relative fluorescence of Nile Red, ordinate) was unchanged at the four
concentrations of control MP. However, in the case of worms exposed to HT-BPL1-IN, there was a
significant 12.5 ± 1.7% reduction (p < 0.001) at the concentration of 1.80 mg/mL and 9.0 ± 1.4%
reduction at the concentration of 3.60 mg/mL (p < 0.001). This preliminary result prompted another
set of experiments through the incubation of fermented baby slurries in the presence of HT-BPL1-IN.
Figure 1. Milk powder supplemented with heat-treated BPL1 (HT-BPL1-IN), reduced fat deposition
in C. elegans measured as fluorescence with red Nile, in a concentration-dependent manner. Worms
were incubated for 3 days with increasing concentrations (abscissa) of milk powder alone (MP) or
milk powder supplemented with HT-BPL1-IN. Data are means ± SD of 120 worms assayed in a
prototype experiment. * p < 0.001 with respect control.
Experiments were done at two concentrations of HT-BPL1, equivalent to 10
8
and 10
9
cells per
reactor (Figure 2). Worms were grown for 3 days in 24-h fermented fecal slurries from two different
infants. The antiobesity drug Orlistat was used as a positive control. In the presence of HT-BPL1, fat
reduction accounted for 17.8 ± 1.1% (p < 0.05) in baby 1 slurry and 20.2 ± 6.0% (p < 0.05) in baby 2. In
Figure 1.
Milk powder supplemented with heat-treated BPL1 (HT-BPL1-IN), reduced fat deposition in
C. elegans measured as fluorescence with red Nile, in a concentration-dependent manner. Worms were
incubated for 3 days with increasing concentrations (abscissa) of milk powder alone (MP) or milk
powder supplemented with HT-BPL1-IN. Data are means
±
SD of 120 worms assayed in a prototype
experiment. * p<0.001 with respect control.
Foods 2020,9, 652 5 of 13
Experiments were done at two concentrations of HT-BPL1, equivalent to 10
8
and 10
9
cells per
reactor (Figure 2). Worms were grown for 3 days in 24-h fermented fecal slurries from two dierent
infants. The antiobesity drug Orlistat was used as a positive control. In the presence of HT-BPL1,
fat reduction accounted for 17.8
±
1.1% (p<0.05) in baby 1 slurry and 20.2
±
6.0% (p<0.05) in baby 2.
In HT-BPL1-IN assays, reductions in fat deposition with respect to control were 21.3
±
1.6% (p<0.05)
in baby 1 and 22.8
±
7.9% in baby 2 (p<0.05). In spite of those pronounced reductions, they did not
reach the level of statistical significance compared to HT-BPL1.
Foods 2020, 9, x FOR PEER REVIEW 10 of 13
HT-BPL1-IN assays, reductions in fat deposition with respect to control were 21.3 ± 1.6% (p < 0.05) in
baby 1 and 22.8 ± 7.9% in baby 2 (p < 0.05). In spite of those pronounced reductions, they did not reach
the level of statistical significance compared to HT-BPL1.
Figure 2. Effects of heat treated BPL1 (HT-BPL1) alone, or added on the infant milk formula Innova
(HT-BPL1-IN), on fat deposition of C. elegans grown for 3 days on the fermented slurries (24 h) from
two different healthy babies. Fat deposition was monitored with relative fluorescence (ordinates) at
the end of the incubation periods, expressed as % reduction of fluorescence compare to control worms
(worms grown on DMSO). Antiobesity drug Orlistat was added to a well at 6 µg/mL as positive
control (DMSO, the solvent for Orlistat). (A, C) experiments done with 108 HT-BPL1 cells/reactor in
slurries from babies 1 (A) and 2 (C). (B, D), experiments done with 109 HT-BPL1/reactor with innova
milk formula (HT-BPL1-IN) or HT-BPL1 alone, in fermented slurries from baby 1 (B) or baby 2 (D).
Data are mean ± SD of 120 worms per well in each condition * p < 0.05, with respect to control. * p <
0.001, with respect to control.
The reduction in fat deposition observed in worms grown in the presence of 109 heat-treated
bacteria was more notable. In all cases, HT-BPL1 reduction was close to Orlistat results. HT-BPL-1
reduced fat deposition by 39.8 ± 5.1% (p < 0.001) and 36.2 ± 1.0% in the presence of HT-BPL1-IN (p <
0.001). In baby 2 slurry, decreased fat deposition was 39.8 ± 4.6% for HT-BPL1, and 37.8 ± 2.7% for
HT-BPL1-IN (p < 0.001 for the two treatments).
3.2. Effects of HT-BPL1, Alone or Included in an Infant Milk Formula, on the Production of Short-Chain
Organic Fatty Acids
This study was aimed at finding out how HT-BPL1 could affect the level of SCFAs and organic
acids generation in fecal slurries from healthy infants, before (0 h) and after fermentation for 24 h.
Table 1 summarizes the results obtained. Figure S1 shows representative chromatograms
obtained in assays 1 and 2. Note that neither propionic acid nor butyric acid underwent significant
changes with respect to control (0 h), both in assay 1 and assay 2. Neither, slurries incubated with
HT-BPL1 alone for 24 h, experimented any change with respect to control, both in the case of acetic
acid or lactic acid. However, when fermentation was done in the presence of HT-BPL1-IN, acetic acid
augmented over 3-fold compared to 0 h samples (9.89 ± 0.02; p < 0.001). The same occurred with lactic
acid that rose from near undetectable levels at 0 h (0.08 ± 0.11 g/L) to 8.34 ± 0.20 g/L (p < 0.001). In
assay 2 these differences were maintained even though in this assay acetate and lactate baseline levels
were higher in the control samples (Table 1). In Figure 3, the net increases of acetate in fermented
fecal slurries, are plotted.
Figure 2.
Eects of heat treated BPL1 (HT-BPL1) alone, or added on the infant milk formula Innova
(HT-BPL1-IN), on fat deposition of C. elegans grown for 3 days on the fermented slurries (24 h) from
two dierent healthy babies. Fat deposition was monitored with relative fluorescence (ordinates) at the
end of the incubation periods, expressed as % reduction of fluorescence compare to control worms
(worms grown on DMSO). Antiobesity drug Orlistat was added to a well at 6
µ
g/mL as positive control
(DMSO, the solvent for Orlistat). (
A
,
C
) experiments done with 10
8
HT-BPL1 cells/reactor in slurries
from babies 1 (
A
) and 2 (
C
). (
B
,
D
), experiments done with 10
9
HT-BPL1/reactor with innova milk
formula (HT-BPL1-IN) or HT-BPL1 alone, in fermented slurries from baby 1 (
B
) or baby 2 (
D
). Data are
mean
±
SD of 120 worms per well in each condition * p<0.05, with respect to control. ** p<0.001, with
respect to control.
The reduction in fat deposition observed in worms grown in the presence of 10
9
heat-treated
bacteria was more notable. In all cases, HT-BPL1 reduction was close to Orlistat results. HT-BPL-1
reduced fat deposition by 39.8
±
5.1% (p<0.001) and 36.2
±
1.0% in the presence of HT-BPL1-IN
(p<0.001). In baby 2 slurry, decreased fat deposition was 39.8
±
4.6% for HT-BPL1, and 37.8
±
2.7% for
HT-BPL1-IN (p<0.001 for the two treatments).
3.2. Eects of HT-BPL1, Alone or Included in an Infant Milk Formula, on the Production of Short-Chain
Organic Fatty Acids
This study was aimed at finding out how HT-BPL1 could aect the level of SCFAs and organic
acids generation in fecal slurries from healthy infants, before (0 h) and after fermentation for 24 h.
Table 1summarizes the results obtained. Figure S1 shows representative chromatograms obtained
in assays 1 and 2. Note that neither propionic acid nor butyric acid underwent significant changes
with respect to control (0 h), both in assay 1 and assay 2. Neither, slurries incubated with HT-BPL1
alone for 24 h, experimented any change with respect to control, both in the case of acetic acid or lactic
acid. However, when fermentation was done in the presence of HT-BPL1-IN, acetic acid augmented
over 3-fold compared to 0 h samples (9.89
±
0.02; p<0.001). The same occurred with lactic acid that
Foods 2020,9, 652 6 of 13
rose from near undetectable levels at 0 h (0.08
±
0.11 g/L) to 8.34
±
0.20 g/L (p<0.001). In assay 2 these
dierences were maintained even though in this assay acetate and lactate baseline levels were higher
in the control samples (Table 1). In Figure 3, the net increases of acetate in fermented fecal slurries,
are plotted.
Table 1.
Levels of short-chain organic fatty acids in slurry fermentations in the absence (control 24 h) or
the presence of HT-BPL1 alone or with milk powder supplemented with HT-BPL1 (HT-BPL1-IN).
Organic Acids Concentration (g/L)
Acetic Acid Lactic Acid Propionic Acid Butyric Acid
Experiment 1
Control (0 h) 2.88 ±0.44 0.08 ±0.11 0.87 ±0.14 1.01 ±0.20
Control (24 h) 3.59 ±0.11 <LOQ 11.22 ±0.10 1.94 ±0.16 *
HT-BPL1 (1 ×108)3.49 ±0.29 <LOQ 11.19 ±0.16 1.90 ±0.24 *
HT-BPL1-IN (1 ×108)9.89 ±0.02 *** 8.34 ±0.20 *** 1.11 ±0.08 0.77 ±0.03
Experiment 2
Control (0 h) 7.98 ±0.27 3.37 ±0.21 0.78 ±0.07 1.00 ±0.07
Control (24 h) 8.18 ±0.13 0.29 ±0.03 * 1.35 ±0.06 3.92 ±0.03 **
HT-BPL1 (1 ×109)7.55 ±0.34 0.10 ±0.05 * 1.32 ±0.28 3.78 ±0.21 **
HT-BPL1-IN (1 ×109) 14.66 ±1.03 ** 10.62 ±0.97 *** 0.85 ±0.01 0.80 ±0.03
Data are means
±
SD of 2 experiments in triplicate; *** p<0.001; ** p<0.01; * p<0.05, with respect control.
1
LOQ
(Limit of Quantification) 0.05 g/L.
Foods 2020, 9, x FOR PEER REVIEW 10 of 13
Table 1. Levels of short-chain organic fatty acids in slurry fermentations in the absence (control 24 h)
or the presence of HT-BPL1 alone or with milk powder supplemented with HT-BPL1 (HT-BPL1-IN).
Organic Acids Concentration (g/L)
Acetic Acid Lactic Acid Propionic Acid Butyric Acid
Experiment 1
Control (0 h) 2.88 ± 0.44 0.08 ± 0.11 0.87 ± 0.14 1.01 ± 0.20
Control (24 h) 3.59 ± 0.11 <LOQ
1
1.22 ± 0.10 1.94 ± 0.16 *
HT-BPL1 (1·10
8
) 3.49 ± 0.29 <LOQ
1
1.19 ± 0.16 1.90 ± 0.24 *
HT-BPL1-IN (1·10
8
) 9.89 ± 0.02 *** 8.34±0.20 *** 1.11 ± 0.08 0.77 ± 0.03
Experiment 2
Control (0 h) 7.98 ± 0.27 3.37 ± 0.21 0.78 ± 0.07 1.00 ± 0.07
Control (24 h) 8.18 ± 0.13 0.29 ± 0.03 * 1.35 ± 0.06 3.92 ± 0.03 **
HT-BPL1 (1·10
9
) 7.55 ± 0.34 0.10 ± 0.05 * 1.32 ± 0.28 3.78 ± 0.21 **
HT-BPL1-IN (1·109) 14.66 ± 1.03 ** 10.62 ± 0.97 *** 0.85 ± 0.01 0.80 ± 0.03
Data are means ± SD of 2 experiments in triplicate; *** p < 0.001; ** p < 0.01; * p < 0.05, with respect
control.
1
LOQ (Limit of Quantification) 0.05 g/L.
Figure 3. Net increase of acetate in fecal baby slurries fermented for 24 h, in the absence (control) and
the presence of HT-BPL1 alone or HT-BPL1 included in an infant milk formula (Innova)
supplemented with HT-BPL1 cells (HT-BPL1-IN). (A) experiment 1 done with a baby slurry in the
presence of 10
8
HT-BPL1 cells; (B), experiment done with a baby slurry in the presence of 10
9
HT-
BPL1 cells. Data calculated from Table 1, by subtracting the background level of acetate in a non-
fermented slurry (0 h in the table).
3.3. Effects of a Milk Powder Supplemented with Heat-Treated BPL1, on Microbiome
Microorganisms present in fecal slurries were identified and quantified in terms of relative
abundance. Diversity by Shannon Index at genus level was evaluated (Supplementary Figure S2),
identifying some differences between babies after 24 h of fermentation in some of the trials, and
showing a later homogeneity once BPL1-HT or BPL1-HT-IN is added, with slight differences between
them.
When we analyzed the potential effect on the regulation of microbiome at genus level in
assays·with 10
8
cells HT-BPL1 incorporated (Figure 4A), we observed that fermentation decreased
the presence of genera Bacteroides and Prevotella, both in majority presence, and increased the
presence of the genus Fecalibacterium and a non-assigned OTU (OTU 94). The comparison of the
results obtained in baby faeces fermented with HT-BPL1 (both HT-BPL1 and HT-BPL1-IN) showed
the highest differences. When comparing HT-BPL1-IN with the rest of the 24 h trials, a trend towards
a higher percentage of the genera Fecalibacterium, Bifidobacterium, Lactobacillus, Flintibacter and OTU94,
whereas lower percentages of Prevotella, Coprococcus and Eubacterium were observed. In the case of
assays with 1·10
9
cells HT-BPL1 per fermentation added (Figure 4B) OTU94, and genera
Fecalibacterium and Flintibacter increased in fecal slurries fermented with HT-BPL1-IN while genera
Eubacterium and Dialister decreased. In the case of Coprococcus, slurry fermentations caused an
elevation of this genus, except in the case of HT-BPL1-IN, with very similar levels to those of fecal
material before being fermented. On the contrary, Akkermansia levels dropped in all fermentations,
except in the case of trials with HT-BPL1-IN assayed.
Figure 3.
Net increase of acetate in fecal baby slurries fermented for 24 h, in the absence (control) and the
presence of HT-BPL1 alone or HT-BPL1 included in an infant milk formula (Innova) supplemented with
HT-BPL1 cells (HT-BPL1-IN). (
A
) experiment 1 done with a baby slurry in the presence of 10
8
HT-BPL1
cells; (
B
), experiment done with a baby slurry in the presence of 10
9
HT-BPL1 cells. Data calculated
from Table 1, by subtracting the background level of acetate in a non-fermented slurry (0 h in the table).
3.3. Eects of a Milk Powder Supplemented with Heat-Treated BPL1, on Microbiome
Microorganisms present in fecal slurries were identified and quantified in terms of relative
abundance. Diversity by Shannon Index at genus level was evaluated (Supplementary Figure S2),
identifying some dierences between babies after 24 h of fermentation in some of the trials, and showing
a later homogeneity once BPL1-HT or BPL1-HT-IN is added, with slight dierences between them.
When we analyzed the potential eect on the regulation of microbiome at genus level in assays
with 10
8
cells HT-BPL1 incorporated (Figure 4A), we observed that fermentation decreased the presence
of genera Bacteroides and Prevotella, both in majority presence, and increased the presence of the genus
Fecalibacterium and a non-assigned OTU (OTU 94). The comparison of the results obtained in baby
faeces fermented with HT-BPL1 (both HT-BPL1 and HT-BPL1-IN) showed the highest dierences.
When comparing HT-BPL1-IN with the rest of the 24 h trials, a trend towards a higher percentage
of the genera Fecalibacterium, Bifidobacterium,Lactobacillus,Flintibacter and OTU94, whereas lower
percentages of Prevotella,Coprococcus and Eubacterium were observed. In the case of assays with
1×109cells
HT-BPL1 per fermentation added (Figure 4B) OTU94, and genera Fecalibacterium and
Flintibacter increased in fecal slurries fermented with HT-BPL1-IN while genera Eubacterium and
Foods 2020,9, 652 7 of 13
Dialister decreased. In the case of Coprococcus, slurry fermentations caused an elevation of this genus,
except in the case of HT-BPL1-IN, with very similar levels to those of fecal material before being
fermented. On the contrary, Akkermansia levels dropped in all fermentations, except in the case of trials
with HT-BPL1-IN assayed.
Foods 2020, 9, x FOR PEER REVIEW 10 of 13
Figure 4. Percentage of identified microorganisms by 16s rRNA by Illumina V3-V4 sequencing. (A)
experiments run with two different baby slurries in the presence of 108 HT-BPL1 cells; (B) experiments
run with two different baby slurries in the presence of 109 HT-BPL1 cells.
4. Discussions
We focused this investigation to find out whether a heat-treated choose Bifidobacterium animalis
subsp. lactis CECT 8145 (BPL1) probiotic preserved some of its functional properties exhibited when
alive, in a fecal model that mimics microbial fermentation in the gut. Initially, this strain showed
activity lowering fat deposition both alive and heat treated in the pre-clinical C. elegans model [19]
0
10
20
30
40
50
60
70
80
Control oh Control 24h HT-BPL1 HT-BPL1-IN
% sequences
OTU_133
OTU_122
OTU_129
Flintibacter
Desul fovib rio
Anaerostipes
Blautia
Gemmiger
Dial ister
Collinsella
Methanobrevib acter
Ruminococcus
Oscillibacter
Faecal ibac terium
Akkermansia
Parabacteroides
Eubacteri um
OTU_137
Coprococcus
Alistipes
No Hit
Prevotella
Bifidobacteriu m
OTU_94
Bacteroid es
0
10
20
30
40
50
60
70
80
90
Control oh Control 24h HT-BPL1 HT-BPL1-IN
% sequences
Flintib acter
Desulfovibrio
Anaerostip es
Blautia
Gemmiger
Dialister
Colli nsella
Methano brevibacte r
Ruminococcus
Osci llibac ter
Faecal ibacterium
Akkermansia
Parabacteroi des
Eubact erium
OTU_137
Copro coccus
Alistipes
No Hi t
Prevotella
Bifidobacterium
OTU _94
Bacteroides
A
B
Figure 4.
Percentage of identified microorganisms by 16s rRNA by Illumina V3-V4 sequencing.
(
A
) experiments run with two dierent baby slurries in the presence of 10
8
HT-BPL1 cells; (
B
)
experiments run with two dierent baby slurries in the presence of 109HT-BPL1 cells.
Foods 2020,9, 652 8 of 13
4. Discussion
We focused this investigation to find out whether a heat-treated choose Bifidobacterium animalis
subsp. lactis CECT 8145 (BPL1) probiotic preserved some of its functional properties exhibited when
alive, in a fecal model that mimics microbial fermentation in the gut. Initially, this strain showed
activity lowering fat deposition both alive and heat treated in the pre-clinical C. elegans model [
19
]
demonstrating the usefulness of the model for probiotic evaluation in fat deposition. After that,
the activity in both formats was validated in two dierent pre-clinical murine models [
20
,
21
] and finally
in a randomized controlled clinical trial done in adult abdominally obese subjects in which the probiotic
validated previous results and showed to improve the anthropometric obesity biomarkers [
27
]. In the
light of these positive outcomes, in this work we attempted to study the eect of strain HT-BPL1 in
infant milk product.
Since the probiotic was to be incorporated into an infant milk, it was initially studied whether this
strain retained its functionality in this matrix. These trials were conducted in the nematode C. elegans.
The small size, short lifespan, complete genetic information and easy maintenance in the laboratory
make C. elegans a valuable animal model to research. It is noteworthy that many of the genes involved
in the synthesis, degradation, and transport of fats are conserved in mammals and have been identified
by RNAi [
28
33
], being its capacity of fat accumulation dependent on the homeostasis of lipogenesis
and lipolysis [
34
]. This nematode conserves 65% of the genes associated with human disease, and in
the case of lipid and energy metabolism, many of the proteins involved in synthesis, degradation
and transporting fats are highly conserved between C. elegans and mammals. Genetic screenings
have identified many gene inactivations that cause fat reduction or fat accumulation [
31
]. This has
led to the use of C. elegans used as a model to evaluate potential obesity therapeutics, and obtain
mechanistic information behind single-gene mutations related to obesity, and define the mechanistic
details of fat metabolism [
28
33
,
35
]. In this sense, compounds targeting conserved pathways regulating
lipids in nematodes are potential successfully candidates for testing in human trials. Trials conducted
in C. elegans model have both confirmed a dose-dependent eect in fat deposition with HT-BPL1
and that HT-BPL1 kept this property when incorporated into the infant milk product (HT-BPL1-IN).
This initial result led us to further develop the model, applying added fecal slurry from the dierent
products to analyze the evolution of the microbiome, the production of organic acids and its eect
on the fat deposition model of C. elegans both with BPL1 alone (HT-BPL1), and with infant milk
(HT-BPL1-IN), in two dierent cell concentrations (10
8
cells/reactor and 10
9
cells/reactor). In an attempt
to “humanize” the intestine of C. elegans, nematodes have been treated with baby fermented fecal
samples and results confirmed its fat-reducing eect in treated nematodes, particularly at 10
9
cells
concentration. These results suggest that microbiome impacts the physiology of the worm, influencing
in health parameters. This is consistent with other C. elegans studies showing host-microbiome
interactions, that are well stablished, like those related with oxidative stress or life-span [
36
]. From the
metabolomic analysis, the results obtained using the fecal slurry model showed that SCFA acetate and
the organic acid lactate were considerably augmented by HT-BPL1-IN, during a 24-h fermentation
period. This augmentation was not observed when HT-BPL1 was added alone, suggesting that either
there may be some kind of synergy between the milk powder and HT-BPL1 or most probably that
the milk powder contains compounds that act on the growth and/or metabolism of dierent acetate
and lactate producing microorganisms in the fermented infant slurries. Infant milk powder contains
lactose which is can be homo-fermented or hetero-fermented by taxons such as Bifidobacterium animalis
subsp. lactis, producing thus lactic and acetic acid respectively [37].
Whatever the mechanism involved, it seems that the increase in acetate has relevant impact in the
context of infant obesity. SCFAs are metabolic products of microbial activity that contribute to intestinal
homeostasis [
38
,
39
]. The most abundant are acetate, propionate and butyrate, that constitute more than
95% of all SCFAs [
38
,
40
]. While acetate and propionate are found in both small and large intestines,
butyrate is mainly present in colon and cecum [
40
]. Acetate also exerts anti-inflammatory activity in
C. elegans via NF-
κ
B [
41
], also presenting functionality as immunomodulator [
42
]. NF-
κ
B signaling is
Foods 2020,9, 652 9 of 13
directly involved in the relationship of inflammation and obesity [
43
]. In a previous work with BPL1
in C. elegans model [
19
], it was observed that this one presented, besides a marked functionality as for
reduction of the corporal fat, antioxidant and antiinflammatory activity. In the context of infant obesity,
some functional observations with acetate deserve a comment. For example, the oral administration of
acetate to mice fed a high-fat diet, reduced body weight, improved insulin sensitivity, decreased total
body fat content and hepatic fat accumulation without changing food intake or physical activity [
44
].
Moreover, oral acetate gavage in an obese and diabetic strain of rats reduced weight gain and improved
glucose tolerance [
44
,
45
]. Furthermore, in mice C-acetate can cross the blood-brain barrier to be taken
up into the hypothalamus that resulted in decreased food intake through appetite suppression [
44
,
45
],
accompanied by increased lactate production [
44
]. There are also some studies in humans, suggesting
that acetate may aect the central regulation of appetite with concomitant reduction of body weight [
44
].
Concomitantly, acetate stimulates leptin secretion from adipocytes to regulate energy balance and
appetite [
46
]. Furthermore, acetate inhibits intracellular lipolysis with reduced lipid overflow and
decreased ectopic accumulation of fat [
44
]. This eect may reduce free fatty acid flux to the liver;
in doing so, acetate decreases fatty liver risk, thus mitigating deterioration of glucose homeostasis [
46
].
Since SCFAs are produced as a result of the bacteria’s own metabolism of the digestive system,
it is to be expected that the greatest changes at the level of bacterial populations would be observed
in the fermented with HT-BPL1-IN assays. So that, microbiome study was done in order to evaluate
how both HT-BPL1 and HT-BPL1-IN impact on bacterial composition. The analysis of the changes
reflects a modulating eect of the microbial populations of INNOVA milk product. Although diversity
varied between dierent samples in the control after 24 h of fermentation probably due to dierent
initial microbial populations that evolve dierentially in the fermentation process, the addition of
HT-BPL1 or HT-BPL1-IN resulted in greater uniformity in diversity within each trial. Among increased
groups, elevated levels of bifidobacteria and lactobacilli can enable the production of lactate and
acetate. In this context, the huge increase in HT-BPL1-IN assays may be produced by the use of milk
carbohydrates such us lactose in organic acids production. Some Bifidobacterium species preferentially
use lactose over glucose as a carbon source when grown in the presence of both sugars [
37
,
47
].
Genus Lactobacillus was in HT-BPL1-IN fermentations although at low percentage levels, being L. gasseri
its representative (data not shown). In the case of this species, there are studies that relate strains of
L. gasseri with regulation of gut environment and function in a postbiotic form [
48
]. Finally, in both
HT-BPL1 concentrations assayed, an increase in Faecalibacterium has been found. This genus is related
to protection against inflammation and related with a healthy human gut microbiome [
49
], so an
increase may have a protective eect against inflammatory dysbiosis and related to this, on metabolic
syndrome, and thus it may be an indicator in this model of incidence in the bacterial microbiome and
modulation of functionality.
The results obtained in this study show a significant eect of the product HT-BPL1-IN on fat
deposition C. elegans model. In previous studies, preclinical obese rodents both in wistar rats fed on a
cafeteria diet [
20
] and with genetically obese rats Zücker rats [
21
] confirmed the results obtained in
C. elegans hat were later observed in humans in a clinical study, randomized double-blind study in
obese patients. All in all, the current study has shown us the eect not only of the probiotic but also of
the product included in its matrix, both in the microbiota and in the fat deposition of C. elegans, which
predict, based on the previous “from worm to human” assays done, a potential eect in humans.
An ongoing clinical trial in infants fed with a milk formula supplemented with heat treated BPL1,
will probably define to what extent this formula is helping to prevent the future development of obesity
in those infants. An increasing number of studies have analyzed the eects of dierent bacterial strains
on SCFA and organic acids production and infant obesity [
50
,
51
]. Our preclinical present study add
evidence in favor of the potential beneficial eects of probiotics and postbiotics on the prevention of
obesity, when added as supplements to milk infant formulas.
Foods 2020,9, 652 10 of 13
5. Conclusions
In conclusion, here we show that heat-treated probiotic strain BPL1, reduces fat deposition in
C. elegans when added to a milk infant formula as a supplement. Although there is an increase in
acetate production, results here reported evidence that other mechanism underlying such eect is
present. The study confirm postbiotic HT-BPL1 eect in milk formula.
Supplementary Materials:
The following are available online at http://www.mdpi.com/2304-8158/9/5/652/s1,
Figure S1: SCFAs and lactate determination by HPLC; Alpha-diversity reflected as Shannon Index; Figure S2:
Alpha-diversity reflected as Shannon Index.
Author Contributions:
Conceptualization, J.M., S.G., D.R., P.M. and E.C.; Methodology, J.F.M.-B. and P.M.;
Validation, A.T., A.G., M.M. and E.C.; Formal analysis, J.F.M.-B., M.M., P.M. and E.C.; Investigation,
Á
.S., N.G.
and V.I.; Data curation, J.F.M.-B. and P.M.; Writing—original draft preparation,
Á
.S., N.G., M.M. and E.C.;
Writing—review and editing, A.G., J.M., S.G., D.R., P.M. and E.C.; Supervision, A.T., A.G., D.R. and P.M.; Project
administration, A.T., S.G., D.R. and P.M.; Funding acquisition, J.M. All authors have read and agreed to the
published version of the manuscript.
Funding: This research was supported by Cátedra UAM-Alter Nutrinfant, Madrid, Spain.
Conflicts of Interest:
A.T., M.M., A.G., declare no conflicts of interest;
Á
.S., N.G., S.G., D.R., P.M. and E.C. are
employees of ADM Biopolis; J.F.M.-B. and V.I. are employees of ADM Lifesequencing; J.M. is employee of Alter
Farmacia SA; The funders had no role in the design of the study; in the collection, analyses, or interpretation of
data; in the writing of the manuscript, or in the decision to publish the results.
References
1.
Abarca-G
ó
mez, L.; Abdeen, Z.A.; Hamid, Z.A.; Abu-Rmeileh, N.M.; Acosta-Cazares, B.; Acuin, C.; Adams, R.J.;
Aekplakorn, W.; Afsana, K.; Aguilar-Salinas, C.A.; et al. Worldwide trends in body-mass index, underweight,
overweight, and obesity from 1975 to 2016: A pooled analysis of 2416 population-based measurement studies
in 128·9 million children, adolescents, and adults. Lancet 2017,390, 2627–2642. [CrossRef]
2.
Kohut, T.; Robbins, J.; Panganiban, J. Update on childhood/adolescent obesity and its sequela. Curr. Opin.
Pediatr. 2019,31, 645–653. [CrossRef] [PubMed]
3.
Oluwagbemigun, K.; Buyken, A.E.; Alexy, U.; Schmid, M.; Herder, C.; Nöthlings, U. Developmental
trajectories of body mass index from childhood into late adolescence and subsequent late adolescence-young
adulthood cardiometabolic risk markers. Cardiovasc. Diabetol. 2019,18, 9. [CrossRef] [PubMed]
4.
Foley, S.; Quinn, S.; Jones, G. Tracking of bone mass from childhood to adolescence and factors that predict
deviation from tracking. Bone 2009,44, 752–757. [CrossRef] [PubMed]
5.
Güngör, N.K. Overweight and obesity in children and adolescents. J. Clin. Res. Pediatr. Endocrinol.
2014
,6,
129–143. [CrossRef] [PubMed]
6.
M
ă
rginean, C.O.; Meli¸t, L.E.; Ghiga, D.V.; M
ă
rginean, M.O. Early inflammatory status related to pediatric
obesity. Front. Pediatr. 2019,7, 241. [CrossRef]
7.
Vael, C.; Verhulst, S.L.; Nelen, V.; Goossens, H.; Desager, K.N. Intestinal microflora and body mass index
during the first three years of life: An observational study. Gut Pathog. 2011,3, 8. [CrossRef]
8.
Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.;
Crowley, J.; Yanagisawa, M.; et al. Eects of the gut microbiota on host adiposity are modulated by the
short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA
2008
,105,
16767–16772. [CrossRef]
9.
Lau, E.; Neves, J.S.; Ferreira-Magalh
ã
es, M.; Carvalho, D.; Freitas, P. Probiotic ingestion, obesity, and
metabolic-related disorders: Results from NHANES, 1999–2014. Nutrients 2019,11, 1482. [CrossRef]
10.
Ahmadi, S.; Jamilian, M.; Karamali, M.; Tajabadi-Ebrahimi, M.; Jafari, P.; Taghizadeh, M.; Memarzadeh, M.R.;
Asemi, Z. Probiotic supplementation and the eects on weight loss, glycaemia and lipid profiles in women
with polycystic ovary syndrome: A randomized, double-blind, placebo-controlled trial. Hum. Fertil.
2017
,
20, 254–261. [CrossRef]
11.
Dror, T.; Dickstein, Y.; Dubourg, G.; Paul, M. Microbiota manipulation for weight change. Microb. Pathog.
2017,106, 146–161. [CrossRef]
Foods 2020,9, 652 11 of 13
12.
Sanchez, M.; Darimont, C.; Panahi, S.; Drapeau, V.; Marette, A.; Taylor, V.H.; Dor
é
, J.; Tremblay, A. Eects of a
diet-based weight-reducing program with probiotic supplementation on satiety eciency, eating behaviour
traits, and psychosocial behaviours in obese individuals. Nutrients 2017,9, 284. [CrossRef] [PubMed]
13.
Adams, C.A. The probiotic paradox: Live and dead cells are biological response modifiers. Nutr. Res. Rev.
2010,23, 37–46. [CrossRef] [PubMed]
14.
Piqu
é
, N.; Berlanga, M.; Miñana-Galbis, D. Health benefits of heat-killed (tyndallized) probiotics: An
overview. Int. J. Mol. Sci. 2019,20, 2534. [CrossRef] [PubMed]
15.
Taverniti, V.; Guglielmetti, S. The immunomodulatory properties of probiotic microorganisms beyond their
viability (ghost probiotics: Proposal of paraprobiotic concept). Genes Nutr.
2011
,6, 261–274. [CrossRef]
[PubMed]
16.
Deshpande, G.; Athalye-Jape, G.; Patole, S. Para-probiotics for preterm neonates-the next frontier. Nutrients
2018,10, 871. [CrossRef]
17.
Vandenplas, Y.; Ludwig, T.; Bouritius, H.; Alliet, P.; Forde, D.; Peeters, S.; Huet, F.; Hourihane, J. Randomised
controlled trial demonstrates that fermented infant formula with short-chain galacto-oligosaccharides and
long-chain fructo-oligosaccharides reduces the incidence of infantile colic. Acta Paediatr.
2017
,106, 1150–1158.
[CrossRef]
18.
Burta, O.; Iacobescu, C.; Mateescu, R.B.; Nicolaie, T.; Tiuca, N.; Pop, C.S. Ecacy and safety of APT036 versus
simethicone in the treatment of functional bloating: A multicentre, randomised, double-blind, parallel group,
clinical study. Transl. Gastroenterol. Hepatol. 2018,3, 72. [CrossRef]
19.
Martorell, P.; Llopis, S.; Gonz
á
lez, N.; Chenoll, E.; L
ó
pez-Carreras, N.; Aleixandre, A.; Chen, Y.; Karoly, E.D.;
Ram
ó
n, D.; Genov
é
s, S. Probiotic strain bifidobacterium animalis subsp. lactis CECT 8145 reduces fat content
and modulates lipid metabolism and antioxidant response in caenorhabditis elegans. J. Agric. Food Chem.
2016,64, 3462–3472. [CrossRef]
20.
Caimari, A.; del Bas, J.M.; Boqu
é
, N.; Crescenti, A.; Puiggr
ò
s, F.; Chenoll, E.; Martorell, P.; Ram
ó
n, D.;
Genov
é
s, S.; Arola, L. Heat-killed Bifidobacterium animalis subsp. Lactis CECT 8145 increases lean mass
and ameliorates metabolic syndrome in cafeteria-fed obese rats. J. Funct. Foods
2017
,38, 251–263. [CrossRef]
21.
Carreras, N.L.; Martorell, P.; Chenoll, E.; Genov
é
s, S.; Ram
ó
n, D.; Aleixandre, A. Anti-obesity properties
of the strain Bifidobacterium animalis subsp. lactis CECT 8145 in Zücker fatty rats. Benef. Microbes
2018
,9,
629–641. [CrossRef] [PubMed]
22.
McBain, A.; Macfarlane, G. Ecological and physiological studies on large intestinal bacteria in relation to
production of hydrolytic and reductive enzymes involved in formation of genotoxic metabolites. J. Med.
Microbiol. 1998,47, 407–416. [CrossRef] [PubMed]
23.
Yuan, S.; Cohen, D.B.; Ravel, J.; Abdo, Z.; Forney, L.J. Evaluation of methods for the extraction and purification
of DNA from the human microbiome. PLoS ONE 2012,7, e33865. [CrossRef] [PubMed]
24.
Klindworth, A.; Pruesse, E.; Schweer, T.; Peplies, J.; Quast, C.; Horn, M.; Glöckner, F.O. Evaluation of general
16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies.
Nucleic Acids Res. 2013,41, e1. [CrossRef]
25.
Marcel, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J.
2011
,
17, 2.
26.
Edgar, R.C.; Haas, B.J.; Clemente, J.C.; Quince, C.; Knight, R. UCHIME improves sensitivity and speed of
chimera detection. Bioinformatics 2011,27, 2194–2200. [CrossRef]
27.
Pedret, A.; Valls, R.M.; Calder
ó
n-P
é
rez, L.; Llaurad
ó
, E.; Companys, J.; Pla-Pag
à
, L.; Moragas, A.;
Mart
í
n-Luj
á
n, F.; Ortega, Y.; Giralt, M.; et al. Eects of daily consumption of the probiotic Bifidobacterium
animalis subsp. lactis CECT 8145 on anthropometric adiposity biomarkers in abdominally obese subjects: A
randomized controlled trial. Int. J. Obes. (Lond.) 2019,43, 1863–1868. [CrossRef]
28.
Ashrafi, K.; Chang, F.Y.; Watts, J.L.; Fraser, A.G.; Kamath, R.S.; Ahringer, J.; Ruvkun, G. Genome-wide RNAi
analysis of Caenorhabditis elegans fat regulatory genes. Nature 2003,421, 268–272. [CrossRef]
29.
Mak, H.Y.; Nelson, L.S.; Basson, M.; Johnson, C.D.; Ruvkun, G. Polygenic control of Caenorhabditis elegans
fat storage. Nat. Genet. 2006,38, 363–368. [CrossRef]
30.
Srinivasan, S.; Sadegh, L.; Elle, I.C.; Christensen, A.G.; Faergeman, N.J.; Ashrafi, K. Serotonin regulates C.
elegans fat and feeding through independent molecular mechanisms. Cell Metab.
2008
,7, 533–544. [CrossRef]
Foods 2020,9, 652 12 of 13
31.
Jones, K.T.; Ashrafi, K. Caenorhabditis elegans as an emerging model for studying the basic biology of
obesity. Dis. Model. Mech. 2009,2, 224–229. [CrossRef] [PubMed]
32.
Shen, P.; Yue, Y.; Park, Y. A living model for obesity and aging research: Caenorhabditis elegans. Crit. Rev.
Food Sci. Nutr. 2018,58, 741–754. [CrossRef]
33.
Martorell, P.; Llopis, S.; Gonz
á
lez, N.; Mont
ó
n, F.; Ortiz, P.; Genov
é
s, S.; Ram
ó
n, D. Caenorhabditis elegans as
a model to study the eectiveness and metabolic targets of dietary supplements used for obesity treatment:
The specific case of a conjugated linoleic acid mixture (Tonalin). J. Agric. Food Chem.
2012
,60, 11071–11079.
[CrossRef]
34.
Shen, P.; Yue, Y.; Zheng, J.; Park, Y. Caenorhabditis elegans: A convenient
in vivo
model for assessing the
impact of food bioactive compounds on obesity, aging, and alzheimer’s disease. Annu. Rev. Food Sci. Technol.
2018,9, 1–22. [CrossRef] [PubMed]
35.
Zheng, J.; Greenway, F.L. Caenorhabditis elegans as a model for obesity research. Int. J. Obes. (Lond.)
2012
,
36, 186–194. [CrossRef] [PubMed]
36.
Zhang, F.; Berg, M.; Dierking, K.; F
é
lix, M.A.; Shapira, M.; Samuel, B.S.; Schulenburg, H. Caenorhabditis
elegans as a Model for Microbiome Research. Front. Microbiol 2017,8, 485. [CrossRef]
37.
Zareba, D.; Ziarno, M.; Obiedzinski, M. Volatile profile of non-fermented milk and milk fermented by
bifidobacterium animalis subsp. lactis. Int. J. Food Prop. 2012,15, 1010–1021. [CrossRef]
38.
Ratajczak, W.; Rył, A.; Mizerski, A.; Walczakiewicz, K.; Sipak, O.; Laszczy ´nska, M. Immunomodulatory
potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim. Pol.
2019
,66, 1–12.
[CrossRef]
39.
Saez-Lara, M.J.; Gomez-Llorente, C.; Plaza-Diaz, J.; Gil, A. The role of probiotic lactic acid bacteria and
bifidobacteria in the prevention and treatment of inflammatory bowel disease and other related diseases:
A systematic review of randomized human clinical trials. Biomed. Res. Int. 2015,2015. [CrossRef]
40.
Sun, M.; Wu, W.; Liu, Z.; Cong, Y. Microbiota metabolite short chain fatty acids, GPCR, and inflammatory
bowel diseases. J. Gastroenterol. 2017,52, 1–8. [CrossRef]
41.
Grompone, G.; Martorell, P.; Llopis, S.; Gonz
á
lez, N.; Genov
é
s, S.; Mulet, A.P.; Fern
á
ndez-Calero, T.;
Tiscornia, I.; Bollati-Fogol
í
n, M.; Chambaud, I.; et al. Anti-inflammatory Lactobacillus rhamnosus CNCM
I-3690 strain protects against oxidative stress and increases lifespan in Caenorhabditis elegans. PLoS ONE
2012,7, e52493. [CrossRef] [PubMed]
42.
Tedelind, S.; Westberg, F.; Kjerrulf, M.; Vidal, A. Anti-inflammatory properties of the short-chain fatty acids
acetate and propionate: A study with relevance to inflammatory bowel disease. World J. Gastroenterol.
2007
,
13, 2826–2832. [CrossRef] [PubMed]
43.
Catrysse, L.; van Loo, G. Inflammation and the metabolic syndrome: The tissue-specific functions of NF-
κ
B.
Trends Cell Biol. 2017,27, 417–429. [CrossRef] [PubMed]
44.
Canfora, E.E.; van der Beek, C.M.; Jocken, J.W.E.; Goossens, G.H.; Holst, J.J.; Olde Damink, S.W.M.;
Lenaerts, K.; Dejong, C.H.C.; Blaak, E.E. Colonic infusions of short-chain fatty acid mixtures promote energy
metabolism in overweight/obese men: A randomized crossover trial. Sci. Rep. 2017,7, 2360. [CrossRef]
45.
Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From dietary fiber to host physiology: Short-chain
fatty acids as key bacterial metabolites. Cell 2016,165, 1332–1345. [CrossRef] [PubMed]
46.
Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on
human metabolism. Gut Microbes 2016,7, 189–200. [CrossRef]
47.
Gonz
á
lez-Rodr
í
guez, I.; Gaspar, P.; S
á
nchez, B.; Gueimonde, M.; Margolles, A.; Neves, A.R. Catabolism of
glucose and lactose in Bifidobacterium animalis subsp. lactis, studied by 13C Nuclear Magnetic Resonance.
Appl. Environ. Microbiol. 2013,79, 7628–7638. [CrossRef]
48.
Sugawara, T.; Sawada, D.; Ishida, Y.; Aihara, K.; Aoki, Y.; Takehara, I.; Takano, K.; Fujiwara, S. Regulatory
eect of paraprobiotic Lactobacillus gasseri CP2305 on gut environment and function. Microb. Ecol. HealthDis.
2016,27, 30259. [CrossRef]
49.
Miquel, S.; Mart
í
n, R.; Rossi, O.; Berm
ú
dez-Humar
á
n, L.G.; Chatel, J.M.; Sokol, H.; Thomas, M.; Wells, J.M.;
Langella, P. Faecalibacterium prausnitzii and human intestinal health. Curr. Opin. Microbiol.
2013
,16,
255–261. [CrossRef]
Foods 2020,9, 652 13 of 13
50.
Balamurugan, R.; George, G.; Kabeerdoss, J.; Hepsiba, J.; Chandragunasekaran, A.M.; Ramakrishna, B.S.
Quantitative dierences in intestinal Faecalibacterium prausnitzii in obese Indian children. Br. J. Nutr.
2010
,
103, 335–338. [CrossRef]
51.
Remely, M.; Hippe, B.; Zanner, J.; Aumueller, E.; Brath, H.; Haslberger, A.G. Gut microbiota of obese,
type 2 diabetic individuals is enriched in faecalibacterium prausnitzii, akkermansia muciniphila and
peptostreptococcus anaerobius after weight loss. Endocr. Metab. Immune Disord. Drug Targets
2016
,16, 99–106.
[CrossRef] [PubMed]
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... This reduction in fat accumulation was also maintained in soy-fermented milk treated with Bifidobacterium animalis subsp. lactis CECT 8145 [31]. Moreover, treatment with this Bifidobacterium strain also induced an improvement of the nematode oxidative stress-response, and an increase in lifespan by 64%. ...
... In this context, a subsequent study by this group demonstrated that an infant milk formula supplemented with heat-treated Bifidobacterium animalis subsp. lactis CECT 8145 (HT-BPL1, as a postbiotic) significantly reduced the fat content in C. elegans, confirming the anti-obesogenic properties of this strain in the nematode [31]. Further investigations demonstrated that lipoteichoic acid (LTA), a postbiotic isolated from the BPL1, was responsible for its lipid-reducing activities, both in NGM plates and glucose-loaded conditions [32]. ...
Article
Full-text available
There is a growing need to develop new approaches to prevent and treat diseases related to metabolic syndromes, including obesity or type 2 diabetes, that focus on the different factors involved in the pathogenesis of these diseases. Due to the role of gut microbiota in the regulation of glucose and insulin homeostasis, probiotics with beneficial properties have emerged as an alternative therapeutic tool to ameliorate metabolic diseases-related disturbances, including fat excess or inflammation. In the last few years, different strains of bacteria, mainly lactic acid bacteria (LAB) and species from the genus Bifidobacterium, have emerged as potential probiotics due to their anti-obesogenic and/or anti-diabetic properties. However, in vivo studies are needed to demonstrate the mechanisms involved in these probiotic features. In this context, Caenorhabditis elegans has emerged as a very powerful simple in vivo model to study the physiological and molecular effects of probiotics with potential applications regarding the different pathologies of metabolic syndrome. This review aims to summarize the main studies describing anti-obesogenic, anti-diabetic, or anti-inflammatory properties of probiotics using C. elegans as an in vivo research model, as well as providing a description of the molecular mechanisms involved in these activities.
... To analyze the differential production of these organic acids, two samples of solubilized agar (one with no S. downii and the other from the inhibition halo generated by this microorganism after culture with S. mutans ATCC 25175 T ) were taken. The organic acids present therein were analyzed by high-performance liquid chromatography coupled to a refractive index detector [54]. The direct action of these acids on S. mutans ATCC 25175 T at supposedly inhibitory concentrations was also studied. ...
Article
Full-text available
Streptococcus downii is a recently reported bacterial species of oral origin, with inhibitory capacity against Streptococcus mutans, Actinomyces naeslundii, Veillonella parvula and Aggregatibacter actinomycetemcomitans, which confers upon it the potential of being an oral probiotic. The aim of the present study was to identify the potential mechanisms by which S. downii exerts its inhibitory effect on S. mutans. To this end, the study assessed the consumption of glucose and proteins available in the culture medium, the modification of the pH, the production of short-chain fatty acids, the changes in the protein panel of the inhibition halo, the production of hydrogen peroxide and the effect of proteinase K. There were no differences in the glucose values or in the protein content of the medium, but there was a reduction in pH (with no effect on the growth of S. mutans). Significant increases were detected in the levels of lactic and formic acid (with no effect on the growth of S. mutans), as well as changes in the peptide panel (with no effect on the growth of S. mutans). The inhibitory effect was maintained in the presence of peroxidase but disappeared after adding proteinase K. Based on these results, it is suggested that the main mechanism of inhibition of S. downii against S. mutans is the production of bacteriocins.
... Paraprobiotics or parabiotics is primarily explored through research conducted on humans and animals (Teame et al. 2020) with limited publications on the development of functional foods (Barros et al. 2021), although the most proficient use of paraprobiotics in the food industry is still in the production of functional foods Siciliano et al. 2021;Á. Silva et al. 2020). There are currently very limited published studies on the use of paraprobiotics for food preservation and packaging purposes . ...
Article
Metabolic by-products are part of the so-called postbiotics of probiotics and other beneficial microorganisms, particularly lactic acid bacteria, which have gained popularity as a feasible alternative to improving food quality and safety. Postbiotics in dry and liquid forms can be easily integrated into food formulations and packaging materials, exhibiting antimicrobial and antioxidant effects owing to the presence of multiple antimicrobials, such as organic acids, bacteriocins, exopolysaccharides and bioactive peptides. Postbiotics can thus control the growth of pathogens and spoilage microorganisms, thereby extending the shelf life of food products. Because of their ability to be easily manufactured without requiring extensive processing, postbiotics are regarded as a safer and more sustainable alternative to synthetic preservatives, which can have negative environmental consequences. Additionally, food manufacturers can readily adopt postbiotics in food formulations without significant modifications. This systematic review provides an in-depth analysis of studies on the use of postbiotics in the biopreservation and packaging of a wide range of food products. The review evaluates and discusses the types of microorganisms, postbiotics preparation and modification techniques, methods of usage in dairy products, meat, poultry, seafood, fruits, vegetables, bread, and egg, and their effects on food quality and safety.
... In particular, the probiotic strain Bifidobacterium animalis subsp. lactis reduces fat mass in the visceral adipose tissue of individuals with obesity [17][18][19], and its inactivated form (postbiotic) has been shown to modulate the gut microbiota composition [20]. Furthermore, studies have shown that increasing the use of probiotics can help to prevent several chronic infant diseases, such as necrotizing enterocolitis and atopic eczema, and improve short-and longterm health [21,22]. ...
Article
Full-text available
Exclusive breastfeeding is highly recommended for infants for at least the first six months of life. However, for some mothers, it may be difficult or even impossible to do so. This can lead to disturbances in the gut microbiota, which in turn may be related to a higher incidence of acute infectious diseases. Here, we aimed to evaluate whether a novel starting formula versus a standard formula provides a gut microbiota composition more similar to that of breastfed infants in the first 6 months of life. Two hundred and ten infants (70/group) were enrolled in the study and completed the intervention until 12 months of age. For the intervention period, infants were divided into three groups: Group 1 received formula 1 (INN) with a lower amount of protein, a proportion of casein to whey protein ratio of about 70/30 by increasing the content of α-lactalbumin, and with double the amount of docosahexaenoic acid/arachidonic acid than the standard formula; INN also contained a thermally inactivated postbiotic (Bifidobacterium animalis subsp. lactis). Group 2 received the standard formula (STD) and the third group was exclusively breastfed (BF) for exploratory analysis. During the study, visits were made at 21 days, 2, 4, and 6 months of age, with ±3 days for the visit at 21 days of age, ±1 week for the visit at 2 months, and ±2 weeks for the others. Here, we reveal how consuming the INN formula promotes a similar gut microbiota composition to those infants that were breastfed in terms of richness and diversity, genera, such as Bacteroides, Bifidobacterium, Clostridium, and Lactobacillus, and calprotectin and short-chain fatty acid levels at 21 days, 2 and 6 months. Furthermore, we observed that the major bacteria metabolic pathways were more alike between the INN formula and BF groups compared to the STD formula group. Therefore, we assume that consumption of the novel INN formula might improve gut microbiota composition, promoting a healthier intestinal microbiota more similar to that of an infant who receives exclusively human milk.
... Moreover, daily consumption of the living strain BPL1 TM ameliorates several anthropometric adiposity biomarkers in abdominally obese adults [41]. Likewise, an infant formula supplemented with BPL1 TM HT reduced fat deposition in C. elegans and augmented acetate and lactate in a fermented infant slurry [42]. ...
Article
Full-text available
Exclusive breastfeeding is recommended for the first six months of life to promote adequate infant growth and development, and to reduce infant morbidity and mortality. However, whenever some mothers are not able to breastfeed their infants, infant formulas mimicking human milk are needed, and the safety and efficacy of each formula should be tested. Here, we report the results of a multicenter, randomized, blinded, controlled clinical trial that aimed to evaluate a novel starting formula on weight gain and body composition of infants up to 6 and 12 months, as well as safety and tolerability. For the intervention period, infants were divided into three groups: group 1 received formula 1 (Nutribén® Innova 1 (Alter Farmacia S.A., Madrid, Spain or INN (n = 70)), with a lower amount of protein, a lower casein to whey protein ratio by increasing the content of α-lactalbumin, and a double amount of docosahexaenoic acid/arachidonic acid than the standard formula; it also contained a thermally inactivated postbiotic (Bifidobacterium animalis subsp. lactis, BPL1TM HT). Group 2 received the standard formula or formula 2 (Nutriben® Natal (Alter Farmacia S.A., Madrid, Spain) or STD (n = 70)) and the third group was exclusively breastfed for exploratory analysis and used as a reference (BFD group (n = 70)). During the study, visits were made at 21 days and 2, 4, 6, and 12 months of age. Weight gain was higher in both formula groups than in the BFD group at 6 and 12 months, whereas no differences were found between STD and INN groups either at 6 or at 12 months. Likewise, body mass index was higher in infants fed the two formulas compared with the BFD group. Regarding body composition, length, head circumference and tricipital/subscapular skinfolds were alike between groups. The INN formula was considered safe as weight gain and body composition were within the normal limits, according to WHO standards. The BFD group exhibited more liquid consistency in the stools compared to both formula groups. All groups showed similar digestive tolerance and infant behavior. However, a higher frequency of gastrointestinal symptoms was reported by the STD formula group (n = 291), followed by the INN formula (n = 282), and the BFD groups (n = 227). There were fewer respiratory, thoracic, and mediastinal disorders among BFD children. Additionally, infants receiving the INN formula experienced significantly fewer general disorders and disturbances than those receiving the STD formula. Indeed, atopic dermatitis, bronchitis, and bronchiolitis were significantly more prevalent among infants who were fed the STD formula compared to those fed the INN formula or breastfed. To evaluate whether there were significant differences between formula treatments, beyond growth parameters, it would seem necessary to examine more precise health biomarkers and to carry out long-term longitudinal studies.
Article
Obesity has become a global public health problem that seriously affects the quality of life. As an important part of human diet, dairy products contain a large number of nutrients that are essential for maintaining human health, such as proteins, peptides, lipids, vitamins, and minerals. A growing number of epidemiological investigations provide strong evidence on dairy interventions for weight loss in overweight/obese populations. Therefore, this paper outlines the relationship between the consumption of different dairy products and obesity and related metabolic diseases. In addition, we dive into the mechanisms related to the regulation of glucose and lipid metabolism by functional components in dairy products and the interaction with gut microbes. Lastly, the role of dairy products on obesity of children and adolescents is revisited. We conclude that whole dairy products exert more beneficial effect than single milk constituent on alleviating obesity and that dairy matrix has important implications for metabolic health.
Article
Full-text available
The World Health Organization recommends exclusive breastfeeding on demand until at least the sixth month of life. Breast milk or infant formula is the infant’s primary food source until the age of one year, followed by the gradual introduction of other foods. During weaning, the intestinal microbiota evolves to a profile close to that of the adult, and its disruption can result in an increased incidence of acute infectious diseases. We aimed to determine whether a novel starting formula (INN) provides gut microbiota compositions more similar to those of breastfed (BF) infants from 6 to 12 months of age compared to a standard formula (STD). This study included 210 infants (70 per group) who completed the intervention until they reached the age of 12 months. In the intervention period, infants were divided into three groups. Group 1 received an INN formula with a lower protein content, a casein to whey protein ratio of approximately 70/30, twice as much docosahexaenoic acid as the STD formula, a thermally inactivated postbiotic (Bifidobacterium animalis subsp. lactis, BPL1TM HT), and twice as much arachidonic acid as the STD formula contained. The second group received the STD formula, while the third group was exclusively BF for exploratory purposes. In the course of the study, visits were conducted at 6 months and 12 months of age. Compared to the BF and STD groups, the Bacillota phylum levels in the INN group were significantly reduced after 6 months. At the end of 6 months, the alpha diversity indices of the BF and INN groups differed significantly from those of the STD group. At 12 months, the Verrucomicrobiota phylum levels in the STD group were significantly lower than those in the BF and INN groups. Based on the comparison between 6 and 12 months, the Bacteroidota phylum levels in the BF group were significantly higher than those in the INN and STD groups. When comparing the INN group with the BF and STD groups, Clostridium sensu stricto 1 was significantly higher in the INN group. The STD group had higher levels of calprotectin than the INN and BF groups at 6 months. The immunoglobulin A levels in the STD group were significantly lower than those in the INN and BF groups after 6 months. Both formulas had significantly higher levels of propionic acid than the BF group at 6 months. At 6 months, the STD group showed a higher quantification of all metabolic pathways than the BF group. The INN formula group exhibited similar behavior to the BF group, except for the superpathway of phospholipid biosynthesis (E. coli). We hypothesize that the novel INN formula may promote an intestinal microbiota that is more similar to the microbiota of an infant who consumes only human milk before the weaning period.
Article
Diverse models have been long explored to impel the progress of food science, while Caenorhabditis elegans (C. elegans) models applied in food toxicology and food function evaluation received vigorous advances recently. With multifaceted advantages such as short lifespan, apparent phenotypes and well-recognized genetic background, C. elegans is becoming a driving model organism in food science. In this review, we comprehensively reviewed the research progress of C. elegans models based on the literature published in past two decades, and systematically categorized the methods of C. elegans models’ construction, summarized the applications of nematodes in food toxicology, mainly involved in microbial contamination, food additives, pesticide residues, heavy metals, food packaging, and physical pollution. C. elegans models’ exploitations for functional foods assessments were also exemplified, and biological properties included anti-oxidant, anti-aging, anti-obesity, anti-glucotoxicity, anti-neurodegenerative and immunoregulation. Research gaps between present shortcomings and future orientations have been further discussed. We believed that this review would inspire ideas and strategies for further nematodes model’s exploration and multi-models-cooperation in food science.
Article
Pulmonary inflammation as one of the extraintestinal manifestations of ulcerative colitis (UC) has attracted extensive attention, and its pathogenesis is closely related to gut dysbiosis. Bifidobacterium animalis subsp. lactis BL-99 (BL-99) can alleviate osteoporosis caused by UC, but less research has been done on other extraintestinal manifestations (EIM) caused by UC. This study aimed to explore the role and potential mechanisms of BL-99 on DSS-induced pulmonary complications in colitis mice. The results showed that BL-99 decreased weight loss, disease activity index score, colonic pathology score, and the production of pro-inflammatory cytokines (e.g., TNF-α, IL-1β, and IL-6) in colitis mice. BL-99 also alleviated DSS-induced lung pathological damage by suppressing the infiltration of pro-inflammatory cytokines, inflammatory monocytes, and macrophages. Furthermore, 16S rRNA gene sequencing showed lower abundances of several potentially pathogenic bacteria (e.g., Burkholderia, Shigella, and Clostridium perfringens) and enrichment in specific beneficial bacteria (e.g., Adlercreutzia and Bifidobacterium animalis) in colitis mice with BL-99 treatment. Targeted metabolomics suggested that BL-99 intervention promoted the production of intestinal acetate and butyrate. Finally, we observed that the pulmonary expression of primary acetate and butyrate receptors, including FFAR2, FFAR3, and, GPR109a, was up-regulated in BL-99-treated mice, which negatively correlated with inflammatory monocytes and macrophages. Altogether, these results suggest that BL-99 might be utilized as a probiotic intervention to prevent the incidence of colitis-related lung injury owing to its ability to shape the intestinal microbiota and suppress inflammation.
Article
Full-text available
Gut microbiota dysbiosis has been recognized as having key importance in obesity- and metabolic-related diseases. Although there is increasing evidence of the potential benefits induced by probiotics in metabolic disturbances, there is a lack of large cross-sectional studies to assess population-based prevalence of probiotic intake and metabolic diseases. Our aim was to evaluate the association of probiotic ingestion with obesity, type 2 diabetes, hypertension, and dyslipidemia. A cross-sectional study was designed using data from the National Health and Nutrition Examination Survey (NHANES), 1999–2014. Probiotic ingestion was considered when a subject reported consumption of yogurt or a probiotic supplement during the 24-hour dietary recall or during the Dietary Supplement Use 30-Day questionnaire. We included 38,802 adults and 13.1% reported probiotic ingestion. The prevalence of obesity and hypertension was lower in the probiotic group (obesity-adjusted Odds Ratio (OR): 0.84, 95% CI 0.76–0.92, p < 0.001; hypertension-adjusted OR: 0.79, 95% CI 0.71–0.88, p < 0.001). Accordingly, even after analytic adjustments, body mass index (BMI) was significantly lower in the probiotic group, as were systolic and diastolic blood pressure and triglycerides; high-density lipoprotein (HDL) was significantly higher in the probiotic group for the adjusted model. In this large-scale study, ingestion of probiotic supplements or yogurt was associated with a lower prevalence of obesity and hypertension.
Article
Full-text available
Background: Obese individuals are often in a chronic inflammatory condition due to the malfunction of immune-related activities in the adipose tissue, involving a transient infiltration of neutrophils within the abdominal fat and their binding to adipocytes. Neutrophil to lymphocyte ratio (NLR) and platelet to lymphocyte ratio (PLR) are considered cost-effective markers for the detection of subclinical inflammation. Our study intends to assess the early stages of inflammation associated with overweight and obesity in children. Materials and Methods: We performed a prospective study with 164 children, aged between 5 and 18 years, admitted to a Pediatric Tertiary Hospital in Romania between January 2018 and January 2019. The patients were divided according to body mass index (BMI) into two groups: Group 1: 77 overweight and obese children (BMI percentile ≥85), and Group 2: 87 children with a normal BMI, in order to evaluate the correlation between BMI and laboratory parameters (CBC, ESR, transaminase, total protein, albumin, and blood glucose levels), inflammatory biomarkers, NLR and PLR, and changes in abdominal ultrasound findings. Results: We found that the leukocyte, lymphocyte, erythrocyte, platelet, CRP, and transaminase levels were significantly higher in the overweight/obese group (p = 0.0379, p = 0.0002, p = 0.0003, p = 0.0006, p < 0.0001, p = 0.0332, and p < 0.0001, respectively). No significant statistical differences between the two groups in terms of neutrophil, hemoglobin, albumin, total protein, and glycemia levels were noted (p > 0.05). Moreover, NLR and PLR did not differ significantly between the two groups (p = 0.4674 and p = 0.9973, respectively). Conclusions: Obesity is associated with systemic low-grade inflammation which is reaching alarming rates worldwide among both children and adults. Our study proved that leukocyte, lymphocyte, erythrocyte, and platelet levels are significantly higher in overweight/obese children, emphasizing the inflammatory status related to this condition. Therefore, obesity-related studies involving pediatric patients are of major interest in order to develop appropriate methods to prevent the development of further complications in adulthood.
Article
Full-text available
Nowadays, the oral use of probiotics is widespread. However, the safety profile with the use of live probiotics is still a matter of debate. Main risks include: Cases of systemic infections due to translocation, particularly in vulnerable patients and pediatric populations; acquisition of antibiotic resistance genes; or interference with gut colonization in neonates. To avoid these risks, there is an increasing interest in non-viable microorganisms or microbial cell extracts to be used as probiotics, mainly heat-killed (including tyndallized) probiotic bacteria (lactic acid bacteria and bifidobacteria). Heat-treated probiotic cells, cell-free supernatants, and purified key components are able to confer beneficial effects, mainly immunomodulatory effects, protection against enteropathogens, and maintenance of intestinal barrier integrity. At the clinical level, products containing tyndallized probiotic strains have had a role in gastrointestinal diseases, including bloating and infantile coli—in combination with mucosal protectors—and diarrhea. Heat-inactivated probiotics could also have a role in the management of dermatological or respiratory allergic diseases. The reviewed data indicate that heat-killed bacteria or their fractions or purified components have key probiotic effects, with advantages versus live probiotics (mainly their safety profile), positioning them as interesting strategies for the management of common prevalent conditions in a wide variety of patients´ characteristics.
Article
Full-text available
Intestinal microbiota is an element of the bacterial ecosystem in all mammalian organisms. These microorganisms play a very important part in the development, functioning, and modulation of the immune system from the moment of birth. In recent years, owing to the use of modern sequencing techniques, the microbiome composition in healthy people has been identified based on bacterial 16S rRNA analysis. Currently, more and more attention is being given to the influence of microorganisms on the host’s cellular metabolism. Analysis of microbial metabolites, among them short-chain fatty acids (SCFAs), and disruption of intestinal microbiota homeostasis in terms of their effects on molecular regulatory mechanisms of immune reactions will surely improve the understanding of the etiology of many common diseases. SCFAs, mainly butyrate, propionate, and acetate, occur in specific amounts, and their proportions can change, depending on the diet, age and diseases. The levels of SCFAs are substantially influenced by the ratio of commensal intestinal bacteria, the disturbance of which (dysbiosis) can lead to a disproportion between the SCFAs produced. SCFAs are regarded as mediators in the communication between the intestinal microbiome and the immune system. The signal they produce is transferred, among others, in immune cells via free fatty acid receptors (FFARs), which belong to the family of G protein-coupled receptors (GPCRs). It has been also confirmed that SCFAs inhibit the activity of histone deacetylase (HDAC) – an enzyme involved in post-translational modifications, namely the process of deacetylation and, what is new, the process of histone crotonylation. These properties of SCFAs have an effect on their immunomodulatory potential i.e. maintaining the anti/pro-inflammatory balance. SCFAs act not only locally in the intestines colonized by commensal bacteria, but also influence the intestinal immune cells, and modulate immune response by multi-protein inflammasome complexes. SCFAs have been confirmed to contribute to the maintenance of the immune homeostasis of the urinary system (kidneys), respiratory system (lungs), central nervous system, and the sight organ.
Article
Full-text available
Background: Reports on body mass index (BMI) trajectories from childhood into late adolescence, their determinants, and subsequent cardiometabolic risk markers, particularly among European populations have been few. Moreover, sex-specifc investigation is necessary considering the sex diference in BMI, and the sex-specifc association between BMI and some cardiometabolic risk markers. Methods: Using a sample from the DOrtmund Nutritional and Anthropometric Longitudinally Designed study, we explored sex-specifc trajectories of the BMI standard deviation score (SDS) from 4 to 18 years of age in 354 males and 335 females by latent (class) growth models. The determinants of trajectory were assessed by logistic regression. We identifed cardiometabolic risk markers that were highly associated with BMI SDS trajectory by random forest regression, and fnally we used generalized linear models to investigate diferences in the identifed cardiometabolic risk markers between pairs of trajectories. Results: We observed four: ‘low-normal weight’, ‘mid-normal weight’, ‘high-normal weight’, and ‘overweight’, and three: ‘‘low-normal weight’, ‘mid-normal weight’, and ‘high-normal weight’ trajectories in males and females, respectively. Higher maternal prepregnancy BMI was associated with the ‘overweight’ trajectory, and with ‘high-normal weight’ trajectory in both sexes. In addition, employed mothers and frst-born status were associated with ‘highnormal weight’ trajectory in females. BMI SDS trajectory was associated with high-density lipoprotein-cholesterol and interleukin-18 (IL-18) in males, and diastolic blood pressure and interleukin-6 (IL-6) in females. However, only males following the ‘overweight’ trajectory had signifcantly higher IL-18 when compared to their ‘low-normal weight’ counterpart. Conclusions: We identifed sex-specifc distinct trajectories of BMI SDS from childhood into late adolescence, higher maternal prepregnancy BMI as a common determinant of the ‘high-normal weight’ and ‘overweight’ trajectories, and ‘overweight’ trajectory being associated with elevated IL-18 in late adolescence–young adulthood. This study emphasizes the role of maternal prepregnancy BMI in overweight, and highlights IL-18 as a cardiometabolic signature of overweight across life.
Article
Full-text available
Background: The effects of probiotic Bifidobacterium animalis subsp. lactis CECT 8145 (Ba8145) and those of its heat-killed form (h-k Ba8145) on human anthropometric adiposity biomarkers are unknown. Objective: To assess the effect of Ba8145 and h-k Ba8145 ingestion on anthropometric adiposity biomarkers. Design: Randomized, parallel, double-blind, placebo-controlled trial with abdominally obese individuals. Participants (n = 135) consumed 1 capsule/day containing 1010 colony forming unit (CFU) of Ba8145, 1010 CFU of h-k Ba8145, or placebo (maltodextrin) for 3 months. Results: Ba8145 ingestion decreased waist circumference, waist circumference/height ratio, and Conicity index (P < 0.05) versus its baseline. Changes versus the placebo group reached significance (P < 0.05) after the h-k Ba8145 treatment. Ba8145 decreased the body mass index compared with baseline and placebo group (P < 0.05). The decrease in visceral fat area after Ba8145 treatments reached significance (P < 0.05) only after h-k Ba8145. When analyses by gender were performed, significance remained only for women. Diastolic blood pressure and HOMA index decreased (P < 0.05) after h-k Ba8145. Gut microbiome analyses showed an increase in Akkermansia spp. after Ba8145 treatment, particularly in the live form, which was inversely related to weight (P = 0.003). Conclusions: In abdominally obese individuals, consumption of Ba8145, both as viable and mainly as heat-killed cells, improves anthropometric adiposity biomarkers, particularly in women. An increase in the gut Akkermansia genus appears as a possible mechanism involved. Our results support Ba8145 probiotic as a complementary strategy in obesity management.
Article
Full-text available
Current evidence supports the use of probiotics in preterm neonates for prevention of necrotizing enterocolitis, mortality and late onset sepsis. Despite the strong evidence, the uptake of this intervention has not been universal due to concerns including probiotic sepsis, pro-inflammatory response and transmission of antibiotic resistance. Critically ill extremely preterm neonates with potentially compromised gut integrity are at higher risk of probiotic sepsis due to translocation. In most countries, probiotics are sold as food supplements with poor quality control. The traditional definition of probiotics as “live microorganisms” has been challenged as many experts have questioned the importance of viability in the context of the beneficial effects of probiotics. Paraprobiotics (ghost probiotics), are defined as non-viable microbial cells (intact or broken) or crude cell extracts (i.e., with complex chemical composition), which, when administered (orally or topically) in adequate amounts, confer a benefit on the human or animal consumer. Current evidence indicates that paraprobiotics could be safe alternatives to probiotics in preterm neonates. High-quality pre-clinical and clinical studies including adequately powered randomised controlled trials (RCTs) are warranted in preterm neonates to explore this new frontier.
Article
Purpose of review: We aim to describe current concepts on childhood and adolescent obesity with a strong focus on its sequela. Childhood obesity is a national epidemic with increasing prevalence over the past three decades placing children at increased risk for many serious comorbidities, previously felt to be only adult-specific diseases, making this topic both timely and relevant for general pediatricians as well as for subspecialists. Recent findings: Childhood obesity develops through an interplay of genetics, environment, and behavior. Treatment includes lifestyle modification, and now metabolic and bariatric surgery is more commonly considered in carefully selected adolescents. The off-label use of adjunct medications for weight loss in childhood and adolescent obesity is still in its infancy, but will likely become the next logical step in those with lifestyle modification refractory obesity. Obesity can lead to several comorbidities, which can persist into adulthood potentially shortening the child's lifespan. Summary: Efforts should be focused primarily on reducing childhood and adolescent obesity, and when indicated treating its sequela in effort to reduce future morbidity and mortality in this precious population. VIDEO ABSTRACT: http://links.lww.com/MOP/A36.
Article
Background: Bloating is a common symptom reported by around 16% to 31% of the general population. Functional bloating is diagnosed in patients with recurrent symptoms of bloating who do not meet the diagnostic criteria of irritable bowel syndrome or other functional gastrointestinal disorders. Methods: This double-blind, multicentre, randomised study compared the safety and efficacy of APT036 (xyloglucan plus tyndallized Lactobacillus reuteri and Bifidobacterium brevis; Aprotecol®) and simethicone in treating functional bloating in adults. APT036 or simethicone were administered orally (3 times/day) for 20 consecutive days, with evaluations at baseline, and on Days 2, 10, 20 (end of treatment) and 30 (follow-up visit). The main outcome measure was safety. Efficacy was assessed at each visit by patient-reported symptom severity (Likert scale) and abdominal girth measurement. A hydrogen breath test was performed at baseline and Day 20. Results: Both APT036 (n=54) and simethicone (n=54) were well tolerated by study subjects; no adverse effects were reported with either treatment. Compared with simethicone, APT036 significantly reduced abdominal distension (P=0.008) and flatulence (P=0.010) from baseline to Day 30. The baseline hydrogen breath test confirmed the presence of small intestinal bacterial overgrowth (SIBO) in all subjects. At Day 20, mean hydrogen gas elevation was below the threshold for a diagnosis of SIBO (<12 ppm above basal on glucose administration) in both study arms. Conclusions: Both APT036 and simethicone had good safety profiles but APT036 was superior to simethicone in relieving symptoms of functional bloating.
Article
We evaluated the effect of oral administration of Bifidobacterium animalis subsp. lactis CECT 8145 strain in Zücker fatty rats. The Zücker fatty rats were randomly divided into two groups (n=10 each) and administered either B. animalis subsp. lactis CECT 8145 (1010 cfu/day) suspended in skim milk, or skim milk alone (control group). Each treatment was administered in drinking bottles from week 5 until week 17 of age. A lean Zücker rat group (standard group) was included to provide normal values for the Zücker strain. This group was administered skim milk in the drinking bottle for the same experimental period as Zücker fatty rats. Body weight gain was greater in the fatty control group than in the fatty rats treated daily with B. animalis subsp. lactis CECT 8145. Furthermore, dry and liquid food intake significantly decreased in the treated Zücker fatty group and these rats also showed decreased plasma ghrelin levels as compared with the Zücker fatty control group. B. animalis subsp. lactis CECT 8145 intake also decreased plasma tumour necrosis factor-α (a proinflammatory cytokine) and plasma malondialdehyde (a biomarker of oxidative stress). Moreover, the ratio plasma total cholesterol/plasma cholesterol transported by high-density lipoproteins, considered as an index for cardiovascular disease, also significantly decreased in the Zücker fatty rats treated with B. animalis subsp. lactis CECT 8145. By contrast, this bacterial strain significantly increased plasma adiponectin (an insulin-sensitising adipokine), but did not produce significant effects on triglyceride levels or glucose metabolism biomarkers. Although further research is required to confirm B. animalis subsp. lactis CECT 8145 is an efficient anti-obesity treatment in humans, the results obtained in this study are promising and point to the health and anti-obesity properties of this bacterial strain.