ArticlePDF Available

Synergistic synbiotic containing fructooligosaccharides and Lactobacillus delbrueckii CIDCA 133 alleviates chemotherapy-induced intestinal mucositis in mice

Authors:

Abstract and Figures

Intestinal mucositis is a commonly reported side effect in oncology patients undergoing chemotherapy and radiotherapy. Probiotics, prebiotics, and synbiotics have been investigated as alternative therapeutic approaches against intestinal mucositis due to their well-known anti-inflammatory properties and health benefits to the host. Previous studies showed that the potential probiotic Lactobacillus delbrueckii CIDCA 133 and the prebiotic Fructooligosaccharides (FOS) alleviated the 5-Fluorouracil (5-FU) chemotherapy-induced intestinal mucosa damage. Based on these previous beneficial effects, this work evaluated the anti-inflammatory property of the synbiotic formulation containing L. delbrueckii CIDCA 133 and FOS in mice intestinal mucosa inflammation induced by 5-FU. This work showed that the synbiotic formulation was able to modulate inflammatory parameters, including reduction of cellular inflammatory infiltration, gene expression downregulation of Tlr2, Nfkb1, and Tnf, and upregulation of the immunoregulatory Il10 cytokine, thus protecting the intestinal mucosa from epithelial damage caused by the 5-FU. The synbiotic also improved the epithelial barrier function by upregulating mRNA transcript levels of the short chain fatty acid (SCFA)-associated GPR43 receptor and the occludin tight junction protein, with the subsequent reduction of paracellular intestinal permeability. The data obtained showed that this synbiotic formulation could be a promising adjuvant treatment to be explored against inflammatory damage caused by 5-FU chemotherapy.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
World Journal of Microbiology and Biotechnology (2023) 39:235
https://doi.org/10.1007/s11274-023-03679-0
RESEARCH
Synergistic synbiotic containing fructooligosaccharides
andLactobacillus delbrueckii CIDCA 133 alleviates
chemotherapy‑induced intestinal mucositis inmice
LaísaMacedoTavares1· LuísCláudioLimadeJesus1· VivianeLimaBatista1· FernandaAlvarengaLimaBarroso1·
AndriadosSantosFreitas1· GabrielaMunisCampos1· MoniqueFerraryAmérico1· TalesFernandodaSilva1·
NinaDiasCoelho‑Rocha1· GiovannaAngeliBelo1· MarianaMartinsDrumond2,3· PamelaMancha‑Agresti2,3·
KátiaDuarteVital4· SimoneOdíliaAntunesFernandes4· ValbertNascimentoCardoso4· AlexanderBirbrair5,6,7·
EnioFerreira5· FlavianoSantosMartins8· JulianaGuimarãesLaguna1· VascoAzevedo1
Received: 14 December 2022 / Accepted: 15 June 2023
© The Author(s), under exclusive licence to Springer Nature B.V. 2023
Abstract
Intestinal mucositis is a commonly reported side effect in oncology patients undergoing chemotherapy and radiotherapy. Pro-
biotics, prebiotics, and synbiotics have been investigated as alternative therapeutic approaches against intestinal mucositis due
to their well-known anti-inflammatory properties and health benefits to the host. Previous studies showed that the potential
probiotic Lactobacillus delbrueckii CIDCA 133 and the prebiotic Fructooligosaccharides (FOS) alleviated the 5-Fluorouracil
(5-FU) chemotherapy-induced intestinal mucosa damage. Based on these previous beneficial effects, this work evaluated the
anti-inflammatory property of the synbiotic formulation containing L. delbrueckii CIDCA 133 and FOS in mice intestinal
mucosa inflammation induced by 5-FU. This work showed that the synbiotic formulation was able to modulate inflammatory
parameters, including reduction of cellular inflammatory infiltration, gene expression downregulation of Tlr2, Nfkb1, and
Tnf, and upregulation of the immunoregulatory Il10 cytokine, thus protecting the intestinal mucosa from epithelial damage
caused by the 5-FU. The synbiotic also improved the epithelial barrier function by upregulating mRNA transcript levels of
the short chain fatty acid (SCFA)-associated GPR43 receptor and the occludin tight junction protein, with the subsequent
reduction of paracellular intestinal permeability. The data obtained showed that this synbiotic formulation could be a promis-
ing adjuvant treatment to be explored against inflammatory damage caused by 5-FU chemotherapy.
Keywords Chemotherapy· Intestinal damage· Synbiotic· Immunomodulation· Intestinal barrier
Laísa Macedo Tavares and Luís Cláudio Lima de Jesus have
contributed equally to this work.
* Vasco Azevedo
vascoariston@gmail.com
1 Department ofGenetics, Ecology, andEvolution, Federal
University ofMinas Gerais, BeloHorizonte, Brazil
2 Federal Center forTechnological Education ofMinas Gerais,
Department ofBiological Sciences, BeloHorizonte, Brazil
3 Federal Center forTechnological Education ofMinas
Gerais, Materials Engineering Post- Graduation Program,
BeloHorizonte, Brazil
4 Department ofClinical andToxicological Analysis, Federal
University ofMinas Gerais, BeloHorizonte, Brazil
5 Department ofGeneral Pathology, Federal University
ofMinas Gerais, BeloHorizonte, Brazil
6 Department ofDermatology, University
ofWisconsin-Madison, Madison, WI, USA
7 Department ofRadiology, Columbia University Medical
Center, NewYork, NY, USA
8 Department ofMicrobiology, Federal University ofMinas
Gerais, BeloHorizonte, Brazil
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
235 Page 2 of 14
Introduction
5-Fluorouracil (5-FU) is an antimetabolite drug widely
used for neoplasia treatment, including head, neck, breast,
and colorectal cancers (Longley etal. 2003; Sougiannis
etal. 2021). Due to its non-specificity, many patients
undergoing treatment with 5-FU develop mucositis as
the main oncological therapy adverse effect (Sonis 2004;
Crombie and Longo 2016). Mucositis is a debilitating
condition characterized by an inflammation of the gas-
trointestinal tract (GIT), which leads to symptoms such
as diarrhea, bleeding, malnutrition, electrolytic misbal-
ance, and infections (Sonis 2004; Touchefeu etal. 2014).
These adverse effects occur due to the compromising effect
of chemotherapy on the intestinal mucosa architecture
(Soares etal. 2008; Savassi etal. 2021), and onintestinal
microbiota composition (van Vliet etal. 2010; Pedroso
etal. 2015; Li etal. 2017).
There is currently no clinical procedure recommended
to prevent or treat mucositis. The palliative options are
based only on analgesics and antibiotics, which shortly
alleviate the symptoms (Kostler etal. 2001). Probiotics,
prebiotics, and synbiotic have been explored as therapeutic
approaches to ameliorate mucositis because they improve
intestinal barrier function and promote systemic and
local host immune system responses (Batista etal. 2020).
These beneficial biotics are also able to regulate the dys-
biotic microbiota and provide bioactive compounds (e.g.,
vitamins, peptides, short-chain fatty acids-SCFAs) with
potential benefits to the host health (Trindade etal. 2018;
Chang etal. 2018; Galdino etal. 2018; Yeung etal. 2020;
Carvalho etal. 2021; Savassi etal. 2021).
Among probiotics, the Lactobacillus delbrueckii subsp.
lactis CIDCA 133 (CIDCA 133) strain has been character-
ized as a promising health-promoting microorganism (de
Jesus etal. 2021b). This strain demonstrated an invitro
immunomodulatory response against Citrobacter roden-
tium and Bacillus cereus infections, stimulating phagocy-
tosis and pathogen clearance by eukaryotic cells (Rolny
etal. 2016; Hugo etal. 2017). Invivo tests demonstrated
that this strain in a fermented milk formulation (107 CFU/
mL), administrated continuous feeding, was able to pre-
vent mucosal damage caused by 5-FU in mice, decreasing
inflammatory infiltrate, preventing goblet cell loss, and
reducing intestinal permeability (De Jesus etal. 2019).
Similar results were observed after heat-inactivation of the
strain (109 CFU/mL, via gavage) or by its cell-free super-
natant (CFS) (Batista etal. 2022), demonstrating that pos-
sibly bacteria cell surface components or secreted products
derived from CIDCA 133 are responsible for its anti-
inflammatory properties, as previously predicted through
a probiogenomics study (de Jesus etal. 2021a, b). The
molecular mechanisms associated with CIDCA 133 (107
CFU/mL, continuous feeding) beneficial effects include
inflammatory NF-κB signaling pathway modulation and
upregulation of the immunoregulatory Il10 cytokine and
epithelial barrier markers (mucin 2, claudin 1, zonulin, and
junctional adhesion molecule gene expression) (Barroso
etal. 2022). Additionally, it has been reported that CIDCA
133 (107 CFU/mL, continuous feeding) presented safety
levels for consumption (de Jesus etal. 2021a, b).
Prebiotics are selectively utilized substrates by host
microorganisms conferring a health benefit. Most are non-
digestible oligosaccharides including fructans, such as the
well-known Fructooligosaccharides (FOS) (Gibson etal.
2017; Davani-Davari etal. 2019). Biotherapeutics appli-
cations of FOS (75–240mg) on intestinal inflammatory
conditions (e.g., colitis and intestinal mucositis) have been
reported, such as its ability to increase SCFA production,
reduce inflammatory cell infiltration, improve epithelial bar-
rier function, attenuate the intestinal mucosa damage and
promote immunological and GIT microbiota modulation
(Capitán-Cañadas etal. 2016; Galdino etal. 2018; Liao etal.
2021; Carvalho etal. 2021).
Prebiotics, when associated with probiotic microorgan-
isms, results in synbiotic formulations that confer a health
benefit on the host (Markowiak and Śliżewska 2017). The
synbiotic can be categorized as complementary, composed
of a probiotic combined with a prebiotic, which is designed
to target autochthonous microorganisms; or as synergistic,
in which the substrate is designed to be selectively utilized
by the co-administered microorganism(s) (Swanson etal.
2020). Beneficial effects of putative synbiotic have been
mainly reported for intestinal inflammatory conditions
(Trindade etal. 2018; Sheng etal. 2020), metabolic diseases
(Soleimani etal. 2019), and enteric infections (Pourmasoumi
etal. 2019), among others, with promising beneficial results.
Thus, based on previous reports in which CIDCA 133 and
FOS were individually able to prevent the mucosal inflam-
mation caused by 5-FU, this study investigated the addi-
tive efficacy of synergistic synbiotic containing CIDCA 133
plus FOS on the 5-FU-induced intestinal mucositis in mice
model.
Materials andmethods
Bacterial growth condition
Lactobacillus delbrueckii subsp. lactis CIDCA 133 strain
(culture collection of CIDCA Center from the National Uni-
versity of La Plata, Argentine) was cultured in MRS broth
(de Man, Rogosa Sharp) (Kasvi, São José dos Pinhais, Bra-
zil) at 37°C for 18h, under microanaerobiosis conditions.
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
Page 3 of 14 235
Then, before mice administration, the bacteria concentration
was adjusted to 108 colony-forming units (CFU)/mL.
Fructooligosaccharides (FOS) andsynbiotic
preparation
The prebiotic supplement FOS NewNutrition (Nutratec,
Ribeirão Preto, Brazil) was used at 240mg/animal/day,
according to Galdino etal. (2018). Before mice administra-
tion, the prebiotic was diluted in modified MRS broth [10g
peptone and 10g meat extract (Kasvi, São José dos Pinhais,
Brazil); 5g yeast extract (Sigma-Aldrich, St. Louis, USA);
1 mL Tween 80, 2g ammonium citrate and 5g sodium
acetate (Synth, Diadema, Brazil); 0.05g manganese sulfate
(Dinâmica, Indaiatuba, Brazil), 0.1g magnesium sulfate and
2g monopotassium phosphate (Vetec, Duque de Caxias,
Brazil) diluted in 1000 mL of distilled water; pH 6.5].
To evaluate the CIDCA 133 property to metabolize FOS,
this probiotic strain was grown on a modified MRS broth
supplemented with FOS (2%) at 37°C for 18h under micro-
anaerobiosis conditions, with its growth rate measured by
absorbance at optical density (600nm), and compared to
its growth on commercial MRS broth (Kasvi, São José dos
Pinhais, Brazil), which contains 2% glucose in its formu-
lation. Then, when CIDCA 133 cultures (grown in MRS
or modified MRS) reached 108 CFU/mL, the probiotic and
synbiotic formulation was administrated to mice. CIDCA
133 (108 CFU/mL) dose was determined by growth curves
using counting colony formation-unit (CFU).
Mice trial protocol
The Ethics Committee in the Use of Animals (CEUA-
UFMG, Protocol n º 34/2021) approved all animal experi-
ment protocols. The Bioterism Center (CEBIO) from the
Federal University of Minas Gerais (UFMG, Brazil) sup-
plied the thirty-six male BALB/c mice (6–8 weeks old)
used in this study. The animals were kept in ventilated poly-
carbonate cages at room temperature (25 ± 2°C), with a
12-hour light/dark cycle, and free access to chow and filtered
water for 24h before experiments.
The animals were divided into six groups (n = 6 animals):
Control (NC), Synbiotic (S); mucositis (MUC); and mucosi-
tis treated with CIDCA 133 (BAC), Fructooligosaccharides
(FOS) or Synbiotic (ST), respectively.
Mice from the NC and MUC groups were fed with MRS
broth, whereas the BAC group was fed with CIDCA 133
(108 CFU/mL) previously grown on MRS. Mice from the
FOS group received modified MRS broth containing the
prebiotic FOS (2%). On the other hand, mice from the S
and ST groups were fed with CIDCA 133 (108 CFU/mL)
previously grown on modified MRS broth containing FOS
(2%). All mice were continuously feeding (ad libitum) for
13 days and bottles (100 mL/day) were changed every 24h.
Mice’s body weight was evaluated daily throughout the
experimental period.
For induction of intestinal inflammation, on the 11th day,
mice in the MUC, BAC, FOS, and ST groups received an
intraperitoneal injection of 5-Fluorouracil drug (300mg/
kg) (Fauldfluor®, Libbs, São Paulo, Brazil) (De Jesus etal.
2019). Mice in the control group (NC) received a saline
solution (NaCl 0.9%) (Vetec, Rio de Janeiro, Brazil). After
72h (14th experimental day), all mice were anesthetized with
a ketamine (80mg/kg) and xylazine (16mg/kg) mixture
(Syntec, Tamboré, Brazil) for blood collection, and eutha-
nized by cervical dislocation, with the small intestine being
collected for further analysis (Fig.1a).
Histopathological andmorphometric analysis
The entire small intestine length was measured, and the
ileum section was collected and prepared for histomor-
phology analysis. For this, the tissues were washed with
phosphate-buffered saline (PBS) 0.1M, and the rolls were
prepared and fixed in 10% buffered formaldehyde solution
(Neon, Suzano, Brazil) for 24h. The material was paraff-
ined and sections of 4-µm thickness were mounted on glass
slides and stained with hematoxylin and eosin. A pathologist
performed the histopathological analysis using a modified
histological score previously determined by Howarth etal.
(1996). For each parameter, a score was given according to
the severity of the lesion in the ileum: absent (0), mild (1),
moderate, (2), and severe (3). For analyses of villus heights
and crypt depth, ten images of the ileum section of each ani-
mal were captured by a BX41 optical microscope (Olympus,
Tokyo, Japan), and analyzed with ImageJ 1.51j.8 software
(National Institute of Health, Bethesda, USA).
Myeloperoxidase andeosinophil peroxidase activity
assays
The recruitment of neutrophils and eosinophils to the ileum
mucosa was measured by their respective enzymes activity:
myeloperoxidase (MPO) and eosinophilic peroxidase (EPO)
(Strath etal. 1985; Souza etal. 2000). Briefly, the tissues
were homogenized and submitted to hypotonic lysis, and
processed with three cycles of freeze-thaw in liquid nitrogen,
respectively, followed by the collection of the supernatant
for enzymatic colorimetric assay.
For EPO, the supernatant (75 µL) was added to 75 µL
of o-phenylenediamine 1.5 mM (Sigma-Aldrich, St. Louis,
MO, USA) previously diluted in Tris–HCl 0.075 mM (pH 8)
plus hydrogen peroxide 6.6 mM (Synth, Diadema, Brazil).
The plate was incubated at 20°C for 30min, and the reaction
was stopped with sulfuric acid 1M (Vetec, Rio de Janeiro,
Brazil). For MPO, the supernatant (25 µL) was added to 25
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
235 Page 4 of 14
µL of 3,3,5,5-Tetramethylbenzidine1.6 mM (Sigma-Aldrich,
St. Louis, MO, USA) diluted in dimethyl sulfoxide (DMSO)
(Sigma-Aldrich, St. Louis, MO, USA). After adding 100 µL
of hydrogen peroxide 0.5mM (Synth), the plate was incu-
bated at 37 ºC for 5min and stopped with sulfuric acid 1M.
The enzymatic assays were measured at 450nm (MPO) and
492nm (EPO) on a microplate spectrophotometer (Bio-
Rad 450 model, Bio-Rad Laboratories). The results were
expressed as EPO or MPO arbitrary units/mg of tissue based
on absorbance.
Intestinal secretory IgA (sIgA) levels
To evaluate the sIgA levels, mice’s intestinal contents were
placed in PBS 0.1M (pH 7.2) supplemented with phenyl-
metanossulfonil fluoride 1 µM (PMSF) (Sigma-Aldrich,
St. Louis, MO, USA). Afterward, the samples were centri-
fuged (5000rpm for 30min at 4°C), and the supernatant
was collected and tested by enzyme-linked immunosorbent
assay (ELISA) using goat anti-mouse IgA (A-4789, Sigma-
Aldrich). The assay absorbance was measured at 492nm
and the concentration of sIgA was expressed in µg/mL of
intestinal contents (Barroso etal. 2021b).
Intestinal permeability
On the 14th experimental day, intestinal permeability was
evaluated by determining radioactivity in the mice’s blood
after oral gavage of 0.1 mL of diethylenetriaminepentaacetic
acid (DTPA) solution labeled with 18.5 megabecquerel
(MBq) of technetium-99m (99mTc-DTPA) (Viana etal.
2010; Carvalho etal. 2021). Four hours after the 99mTc-
DTPA administration, the mice’s blood was collected and
the radioactivity was determined in the gamma radiation
counter (Wizard 1480, PerkinElmer/Wallac, Turku, Fin-
land). The results were expressed as the percentage of the
dose injected per gram (% ID/g) of blood, following: [counts
per minute (blood)/(the standard dose)] × 100 (Viana etal.
2010).
Cytokines andepithelial barrier markers gene
expression
The profile of cytokines and genetic markers involved with
epithelial barrier function was evaluated through relative
gene expression. For this, ileum’s total RNA isolation was
carried out using Trizol (Ludwig Biotec, Alvorada, Bra-
zil), according to the manufacturer’s recommendations.
The RNA was evaluated qualitatively and quantitatively in
1.5% agarose gel and NanoDrop® 2000 spectrophotometer
(Thermo Scientific, Waltham, MA, USA), respectively.
Residual genomic DNA was removed using the Turbo DNA-
free™ Kit (Carlsbad, CA, USA). Complementary DNA was
obtained from 2µg of RNA using the High-Capacity cDNA
Reverse Transcription kit (Applied Biosystems™, Ther-
moFisher, Waltham, MA, USA), according to the recom-
mended protocol.
Gene expression analysis was performed using Pow-
erUp™ SYBR® Green Master Mix (ThermoFisher®) on
Applied Biosystems 7900HT Fast Real-Time PCR System
under the following conditions: 95°C for 10min, and 40
cycles of 95°C for 15s and 60°C for 1min. The gene-
specific primers are listed in Table1. All samples were ana-
lyzed in duplicate, and Gapdh (glyceraldehyde 3-phosphate
dehydrogenase) and Actb (actinbeta) genes were used as
endogenous references. Data were analyzed according to
the relative expression using the 2–ΔΔCT method (Livak and
Schmittgen 2001).
Statistical analysis
The Shapiro-Wilk test was used to evaluate data normality.
Data were analyzed using one-way ANOVA followed by
Tukey’s post-test (parametric data) or Kruskal-Wallis’ test
followed by Dunn’s post-test (non-parametric data) using the
GraphPad Prism 8.0 software. Statistically significant find-
ings were marked according to the p < 0.05 cutoffs.
Results
Lactobacillus delbrueckii CIDCA 133 metabolizes FOS
The data in Fig.1b showed that, besides glucose, CIDCA
133 is also able to metabolize the prebiotic FOS as a carbon
source. However, its growth rate was lower (Fig.1b). After
this investigation, the therapeutic effect of this formula-
tion was analyzed in mice with mucositis induced by the
5-Fluorouracil.
5-FU induced body weight loss in the MUC group
(− 16.06 ± 1.80%) compared to the control group (NC)
(0.93 ± 2.10%) (p < 0.05), but no protective effect was
observed on this parameter after treatments with probiotic
CIDCA 133 (BAC), prebiotic FOS (FOS) and synbiotic (ST)
(p > 0.05) (Fig.1c). After 24h of administration, it was pos-
sible to observe that CIDCA 133 reached a maximum count
of 1010 CFU/mL in both MRS and MRS modified medium,
with a pH of 4.13 and 3.93, respectively.
Synbiotic modulates thegene expression ofinflammatory
cytokines
Mice in the MUC group exhibited gene expression upregu-
lation of Tlr2 (p < 0.0001) (Fig.2a) and Nfkb1 (p < 0.0001)
(Fig.2b), the cytokines Tnf (p < 0.0001) (Fig. 2c), Il1b
(p < 0.05) (Fig. 2d) and Tgfb1 (p < 0.001) (Fig. 2g), and
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
Page 5 of 14 235
downregulation of immunoregulatory cytokine Il10
(p < 0.0001) (Fig.2f) compared to the control group (NC).
On the other hand, non-inflamed mice that consumed the
synbiotic (S) presented upregulation of the gene expression
of Il1b (Fig.2d), Il12 (Fig.2e), and Il10 (Fig.2f) cytokines
compared to the control group (p < 0.001).
The treatment with probiotic (BAC) or prebiotic (FOS)
reduced the mRNA expression of Tlr2 (Fig.2a), Nfkb1
(Fig.2b), Tnf (Fig.2c), Il12 (Fig.2e), and Tgfb1 (Fig.2g)
(p < 0.0001). Treatment with prebiotic (FOS) was also
effective to upregulate the transcripts levels of Il10 (Fig.2f)
(p < 0.0001). Synbiotic (ST) downregulated the gene expres-
sion of Tlr2 (Fig.2a), Nfkb1 (Fig.2b), Tnf (Fig.2c), and
Tgfb1 (Fig.2g) (p < 0.0001) and upregulate the transcripts
levels of the immunoregulatory cytokine Il10 (Fig.2f)
(p < 0.0001). Nevertheless, this treatment was more effec-
tive for modulating only the relative gene expression of
Il1b (Fig.2d), Il12 (Fig.2e), and Il10 (Fig.2f) cytokines
(p < 0.001) than probiotic and prebiotic treatments.
Synbiotic contributes tothemaintenance
oftheintestinal epithelial barrier
Mice in the MUC group (0.065 ± 0.01% ID/g) exhibited
increased intestinal permeability (p < 0.001) (Fig. 3a)
and gene expression downregulation of the tight junction
protein occludin (Fig.3b) compared to the control group
(NC) (0.005 ± 0.000095% ID/g) (p < 0.001). Probiotic
(BAC) (0.01 ± 0.0050% ID/g) (p < 0.0001), prebiotic (FOS)
(0.045 ± 0.0083% ID/g) (p < 0.05), and synbiotic (ST)
(0.02 ± 0.01% ID/g) (p < 0.0001) treatments significantly
reduced 5-FU-induced intestinal permeability (Fig.3a).
Additionally, prebiotic (FOS) and synbiotic treatment (ST)
upregulated the gene expression of the tight junction claudin
1 (Fig.3c) compared to the MUC group (p < 0.05). Probiotic
and synbiotic were also effective in upregulating the gene
expression of the GPR43 receptor (Fig.3d). However, the
synbiotic treatment (ST) was more effective for modulating
the occludin gene expression (p < 0.01) (Fig.3c) than the
probiotic and prebiotic individually. No difference in sIgA
levels was observed between the inflamed groups (MUC,
BAC, FOS, and ST) (p > 0.05) (Fig.3e).
Synbiotic reduces cell inflammatory infiltrate
andameliorates theintestinal epithelial
architecture
Histopathological analysis demonstrated that 5-FU induced
significant alterations (e.g., intense polymorphonuclear cell
infiltration, villus shortening, and crypt depth increase)
Table 1 List of primer
sequences used in qPCR. Gene Primer sequence (5……. 3) References
Actb F: GCT GAG AGG GAA ATC GTG CGTG (Volynets etal. 2016)
R: CCA GGG AGG AAG AGG ATG CGG
Gapdh F: TCA CCA CCA TGG AGA AGG C (Giulietti etal. 2001)
R: GCT AAG CAG TTG GTG GTG CA
Tlr2 F: ACA ATA GAG GGA GAC GCC TTT (Chang etal. 2020)
R: AGT GTC TGG TAA GGA TTT CCCAT
Nfkb1 F: GTG GAG GCA TGT TCG GTA GTG (Zheng etal. 2017)
R: TCT TGG CAC AAT CTT TAG GGC
Tnf F: ACG TGG AAC TGG CAG AAG AG (Song etal. 2013)
R: CTC CTC CAC TTG GTG GTT TG
Il1b F: CTC CAT GAG CTT TGT ACA AGG (Song etal. 2013)
R: TGC TGA TGT ACC AGT TGG GG
Il12p40 F: GGA AGC ACG GCA GCA GAA TA (Giulietti etal. 2001)
R: AAC TTG AGG GAG AAG TAG GAA TGG
Il10 F: GGT TGC CAA GCC TTA TCG GA (Giulietti etal. 2001)
R: ACC TGC TCC ACT GCC TTG CT
Tgfb1 F: TGA CGT CAC TGG AGT TGT ACGG (Giulietti etal. 2001)
R: GGT TCA TGT CAT GGA TGG TGC
Ocln F: ACT CCT CCA ATG GAC AAG TG (Volynets etal. 2016)
R: CCC CAC CTG TCG TGT AGT CT
Cldn1 F: TCC TTG CTG AAT CTG AAC A (Volynets etal. 2016)
R: AGC CAT CCA CAT CTT CTG
Gpr43 F: ACA GTG GAG GGG ACC AAG AT (Xu etal. 2019)
R: GGG GAC TCT CTA CTC GGT GA
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
235 Page 6 of 14
in the ileum mucosa compared to the control group (NC)
(Fig.4). These data were correlated with a respective shorter
small bowel length (Fig.5a) and higher histopathological
scores (Fig.5b) (p < 0.05). Non-inflamed mice that con-
sumed the synbiotic (S) showed a normal intestinal mucosa
aspect, evidencing that this biotic did not change the intes-
tinal epithelial architecture (Fig.4).
The synbiotic treatment (ST) increased the small intes-
tine length (p < 0.001) (Fig.5a) and ameliorated the intes-
tinal architecture by improving the villus length (Fig.5c)
(p < 0.0001), the crypt depth (p < 0.0001) (Fig.5d), and the
villus height to crypt depth ratio (p < 0.0001) (Fig.5e). A
protective effect was also observed in BAC and FOS groups
to these morphometric parameters (Fig.4b, c, d, e, f). Nev-
ertheless, the synbiotic treatment (ST) was more effective in
increasing the crypt/villus ratio (Fig.5e) and the small intes-
tine length (Fig.5a) than probiotic or prebiotic treatments.
Regarding polymorphonuclear cells infiltrating into
ileum mucosa, high levels of MPO (neutrophil infil-
trate) (1.83 ± 0.60 AU/mg) (Fig.5f) and EPO (eosinophil
infiltrate) (1.06 ± 0.14 AU/mg) (Fig.5g) enzyme activity
were observed after 5-FU (MUC group) administration com-
pared to the negative control (NC) (MPO: 0.13 ± 0.06 AU/
mg; EPO: 0.71 ± 0.15 AU/mg) (p < 0.01) (Fig.5f, g). After
synbiotic treatment (ST), the activity levels of these enzymes
were reduced (MPO: 1.10 ± 0.28 AU/mg; EPO: 0.065 ± 0.01
AU/mg) (p < 0.05). A protective effect was also observed
for the group treated only with the prebiotic FOS (MPO:
0.94 ± 0.47 AU/mg; EPO: 0.29 ± 0.17 AU/mg) (p < 0.01).
However, the synbiotic treatment (ST) was more effective
in reducing EPO activity levels than probiotic and prebiotic
treatments individually. On the other hand, the BAC treat-
ment did not improve these parameters (p > 0.05) (Fig.5f, g).
Discussion
The beneficial action of synbiotic on intestinal mucosi-
tis has been reported (Smith etal. 2008; Trindade etal.
2018; Savassi etal. 2021). Thus, considering the previous
Fig. 1 Lactobacillus delbrueckii CIDCA 133 metabolizes the prebi-
otic FOS. aExperimental scheme. bGrowth curve of CIDCA 133 in
modified MRS supplemented with FOS (2%). cThe therapeutic effect
of the synbiotic on mice weight loss. Different letters (a, b) indicate
statistically significant differences (p < 0.05) by Kruskal–Wallis fol-
lowed by Dunn’s post-test. NC: control; S: synbiotic; MUC: mucosi-
tis; BAC: mucositis treated with CIDCA 133; FOS: mucositis treated
with Fructooligosaccharides; ST: mucositis treated with synbiotic
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
Page 7 of 14 235
Fig. 2 Synbiotic modulates cytokines gene expression in mice
inflamed with 5-FU. aTlr2, b, Nfkb1, cTnf, dIl1b, eIl12, fIl10, and
gTgfb1. Different letters (a, b, c, d) indicate statistically significant
differences (p < 0.05) by ANOVA followed by Tukey’s post-test. NC:
control; S: synbiotic; MUC: mucositis; BAC: mucositis treated with
CIDCA 133; FOS: mucositis treated with Fructooligosaccharides;
ST: mucositis treated with synbiotic
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
235 Page 8 of 14
beneficial effects of FOS (Galdino etal. 2018; Carvalho
etal. 2021) and L. delbrueckii CIDCA 133 (De Jesus etal.
2019; Barroso etal. 2022; Batista etal. 2022) on mucositis,
this study investigated the anti-inflammatory role of a syn-
biotic containing the association of CIDCA 133 and FOS in
5-FU-induced intestinal mucositis.
Our study shows that L. delbrueckii CIDCA 133, despite
glucose, can also metabolize FOS. However, the growth rate
of CIDCA 133 with FOS was lower than in a glucose-con-
taining medium. Similar results were also observed by (Cao
etal. 2019) when studying the effects of different oligosac-
charides (FOS, GOS, and MOS) and glucose on Lactiplan-
tibacillus plantarum ATCC14917 growth (Cao etal. 2019).
Therefore, based on this finding, although glucose is the
main carbohydrate source that is easily assimilated by most
species of gut microbiota (Moraes etal. 2014), FOS can
be an interesting alternative that can prevent the unwanted
growth of some bacteria and other pathogenic microorgan-
isms that require rich and facile nutritional sources (Pas-
salacqua etal. 2016).
Although some studies report that CIDCA 133 (De Jesus
etal. 2019) and FOS (Galdino etal. 2018) independently
ameliorate intestinal mucositis, in our study neither treat-
ment, including the synbiotic formulation, was effective in
reducing the 5-FU-induced mice weight loss, an important
clinical parameter evaluated on chemotherapy practice.
Similar results were obtained by (Carvalho etal. 2021) and
(Barroso etal. 2022), when using FOS (250mg/day) and
continuous feeding CIDCA 133 (107 CFU/mL). However,
in a fermented milk formulation (De Jesus etal. 2019) and
high concentrations of FOS (550mg/day) (Trindade etal.
2018), was observed that both were able to ameliorate the
mice’s weight loss. Thus, one possible explanation for this
outcome is that the beneficial effects of these biotics, either
Fig. 3 Synbiotic regulates the epithelial barrier. a Intestinal perme-
ability, b occludin, c claudin 1, dGPR43 gene expression, e sIgA
levels. Different letters (a, b, c, d, e) indicate statistically significant
differences (p < 0.05) by ANOVA followed by Tukey’s post-test.NC:
control; S: synbiotic; MUC: mucositis; BAC: mucositis treated with
CIDCA 133; FOS: mucositis treated with Fructooligosaccharides;
ST: mucositis treated with synbiotic
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
Page 9 of 14 235
alone or in combination, depend on FOS concentration and
CIDCA 133 growth matrix type, respectively.
According to the pathobiology of mucositis, the main
histopathological changes associated with chemotherapy-
induced mucositis include villus atrophy, crypt goblet cell
reduction, inflammatory cells infiltrate in the mucosa, and
mucosal barrier dysfunction (Sonis 2004; Dahlgren etal.
2021). Cell damage after 5-FU administration is primar-
ily due to its ability to generate reactive oxygen species
(ROS) and cellular apoptosis, with subsequent production
of inflammatory cytokines derived from the activation of
signaling pathways such as NF-κB (Sonis 2004; Sougiannis
etal. 2021; Dahlgren etal. 2021). The histological analysis
performed in our study revealed that the administration of
synbiotic in inflamed mice ameliorated the intestinal epithe-
lium architecture by improving intestinal villus height, and
the villus/crypt ratio and reducing the polymorphonuclear
cell infiltrate. This ameliorative effect was also observed
macroscopically via restoration of the small bowel length.
In our study, the beneficial effects of synergistic synbiotic
to ameliorate the intestinal epithelium architecture can be
associated with its property to reduce the inflammatory pro-
cess, via inhibition of mRNA transcript levels of pro-inflam-
matory markers (e.g., Tlr2, Nfkb1, and Tnf) and upregulation
of the immunoregulatory cytokine Il10, which is essential
to decreasing the intensity of inflammatory response at the
tissue damage site (Ihara etal. 2017; Wei etal. 2020). This
process could reduce the recruitment of inflammatory cells
and the production of pro-inflammatory cytokines in the
intestinal mucosa, thus restoring intestinal homeostasis.
Other studies support these findings, in which the adminis-
tration of different synbiotic formulations alleviated chem-
otherapy-associated mucosal damage and other GIT-related
inflammatory conditions by improving the intestinal inflam-
matory process and reducing neutrophils recruitment and
secretion of pro-inflammatory cytokines (e.g., IL1b, IL17A,
TNF) (Smith etal. 2008; Trindade etal. 2018; Sheng etal.
2020; Savassi etal. 2021).
Our study also showed that 5-FU induced upregulation
of Tgfb1 and downregulation of Il10 gene expression, cor-
roborating previous studies (Barbosa etal. 2018; Wu etal.
2020; Al-Khrashi etal. 2022; Barroso etal. 2022; Batista
etal. 2022). Synbiotic treatment modulated this profile,
upregulating Il10 and downregulating Tgfb1 cytokines gene
expression. Both cytokines act as anti-inflammatory agents
(Ihara etal. 2017; Wei etal. 2020). Thus, we believe that
the high transcript levels of the TGFβ1 cytokine can suggest
a compensatory anti-inflammatory mechanism to restore
gut homeostasis disrupted by 5-FU chemotherapy, due to
reduced levels of immunoregulatory IL10 cytokine (Gomes-
Santos etal. 2017; Batista etal. 2022).
It is also important to highlight that the transcripts levels
of IL1b and IL12 cytokines and high levels of sIgA were
increased after synbiotic treatment. In general, some studies
Fig. 4 Synbiotic restores intestinal mucosa architecture altered by 5-FU
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
235 Page 10 of 14
of chemotherapy-induced intestinal mucositis recognize
these cytokines as pro-inflammatory (Yeung etal. 2015; Li
etal. 2017; Shen etal. 2021; Quintanilha etal. 2022; Coe-
lho-Rocha etal. 2022). However, the gut secretion of IL1b
can be associated with intestinal barrier repair (Wu etal.
2022) or induction of IgA production (Jung etal. 2015).
On the other hand, IL12 secretion can promote the activa-
tion of natural killer (NK) cells and the development of Th1
cells, thus increasing the immune defense against infections
(Shida etal. 2011). Thereby, a possible explanation for the
synergistic synbiotic increased mRNA transcript of IL1b and
IL12, and sIgA levels can be linked to an immunostimula-
tory mechanism of this biotic to control pathogenic bacteria-
causing infections resulting from the dysbiosis induced by
5-FU, which in our study can be associated with high mRNA
transcript levels of TLR2. This receptor recognizes bacteria
factors as being important to the communication between
microbes and the gut (van Vliet etal. 2010).
Chemotherapy-induced intestinal damage seems to be
related to TLR2 activation (Wu etal. 2020; Wei etal. 2021).
A previous study of our research group demonstrated that
5-FU (300mg/Kg) causes alterations in the microbiota
composition (Carvalho etal. 2018). This information was
also corroborated by Andrade etal. (2023). On the other
hand, our research group has reported that this 5-FU dose
increases the gene expression of TLR2 (Barroso etal. 2021,
2022; Batista etal. 2022). Based on this information, even
though we have not directly evaluated dysbiosis in our work,
Fig. 5 Synbiotic reduces epithelial damage induced by 5-FU.aSmall
bowel length. b Histological score. c Villus length. d Crypt
depth. e Villus/crypt ratio. (f) MPO activity (g) EPO activity. Dif-
ferent letters (a, b, c, d) indicate statistically significant differences
(p < 0.05) by Kruskal–Wallis followed by Dunn’s post-test or by
ANOVA followed by Tukey’s post-test. NC: control; S: synbiotic;
MUC: mucositis; BAC: mucositis treated with CIDCA 133; FOS:
mucositis treated with Fructooligosaccharides; ST: mucositis treated
with synbiotic
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
Page 11 of 14 235
evidence shows that this dose of 5-FU causes dysbiosis and
TLR alterations, suggesting that modulation of this receptor
may be evidence of microbiota regulation by the synbiotic
formulation. Thus, although the pathogen inhibition prop-
erty by CIDCA 133 (Rolny etal. 2016; Hugo etal. 2017)
and increased number of beneficial bacteria (e.g., SCFA-
producer Bifidobacterium) after FOS supplementation (Mao
etal. 2018; Dou etal. 2022) have been reported, we rein-
force that further studies should be conducted to evaluate the
impact of the synbiotic containing FOS plus CIDCA 133 on
microbiota regulation.
Another common feature of chemotherapy-induced
mucositis is intestinal barrier disruption due to the apoptosis
of intestinal epithelial cells, resulting in an intestinal perme-
ability increase (Nakao etal. 2012; Song etal. 2013; Wardill
etal. 2014; Li etal. 2017). Our study observed that the syn-
biotic treatment ameliorated the integrity of the epithelial
barrier disrupted by 5-FU administration via upregulation
of tight junctions’ occludin and claudin 1 gene expression
and reduction of intestinal permeability. These results are
consistent with the data obtained by(Barroso etal. 2022)
and (Carvalho etal. 2021). These authors demonstrated that
increased gene expression of tight junctions (e.g., claudin 1,
occludin, zonulin) following administration of CIDCA 133
or FOS, respectively, was associated with a reduction of the
intestinal permeability and mitigation of epithelial damage
induced by 5-FU chemotherapy (Carvalho etal. 2021; Bar-
roso etal. 2022).
The gut barrier integrity and immunomodulatory activ-
ity observed in synbiotic treatment can be also linked to
CIDCA 133 metabolism-derived SCFA production, evalu-
ated in our study through the upregulation of the Gpr43
receptor gene expression. SCFA, such as acetate, propion-
ate, and butyrate, are metabolites produced through the fer-
mentation of dietary substrates by commensal microbiota.
These compounds, mainly acetate, can improve the epithelial
barrier function through the induction of genes encoding
tight-junctions (TJ) components and the production of anti-
microbial peptides (AMPs) (e.g., RegIIIγ and β-defensins)
by intestinal epithelial cells (IECs) (Parada Venegas etal.
2019a). Regarding immunomodulation properties, their anti-
inflammatory effects in intestinal mucosa are mainly derived
from their ability to activate their specific cell surface G-pro-
tein coupled receptors (GPCR) (e.g., GPR41, GPR43, and
GPR109A) present in immune cells and intestinal epithelial
cells (Rooks and Garrett 2016; Parada Venegas etal. 2019;
Russo etal. 2019).
The beneficial impact of SCFA-producing beneficial
bacteria on intestinal inflammation has been reported. In
this context, the potential probiotic Lacticaseibacillus
rhamnosus improved inflammatory damage of the intestinal
mucosa affected by 5-FU chemotherapy by reducing NLRP3
inflammasome and enhancing the expressions of ZO-1 and
Occludin in the small intestinal mucosa (Yue etal. 2022).
These ameliorative effects were associated with Lb. rham-
nosus influence to increase the SCFA (acetate, propionate,
and butyrate) levels in mice stool (Yue etal. 2022). Similar
results were also reported for Streptococcus thermophilus
ST4 (Shen etal. 2021), which alleviated 5-FU-induced epi-
thelial damage by reducing serum levels of TNF-α, IL-1β,
and IL-6 cytokines and increasing acetic acid levels in mice
stool. Thus, we believed that the production and interaction
of SCFA-derived from CIDCA 133 metabolism with the
GPR43 receptor may have promoted the anti-inflammatory
IL10 cytokine-producing cell activation, with subsequent
suppression of pro-inflammatory markers, such as TNFα,
derived from NF-κB signaling pathway activation, being a
possible mechanism used by the synbiotic formulation to
alleviated mucositis.
Our study has some limitations that should be improved
in future related work. Firstly, our work was conducted using
a laboratory culture broth (MRS) and a continuous feed-
ing (ad libitum) route. Thus, the results may be different
when using other growth matrices of CIDCA 133 (e.g., fer-
mented milk formulation) and treatment administration route
(gavage). In addition, with continuous feeding, we have no
control over exactly how much bacteria the mice consume
throughout the day. Secondly, another aspect that deserves
attention is that if we consider translating the approach to
humans, it is important to point out that they do not con-
sume biotic formulation (probiotics, prebiotics, synbiotics)
or MRS as the only hydric source throughout the day. Fur-
thermore, comparing the effect of the synbiotic formulation
to the effects of the prebiotic and probiotic alone deserves
attention, since the probiotic was grown on another carbon
source (such as glucose), which could lead to difficulties in
interpreting the results in a continuous feeding administra-
tion. Finally, the molecular mechanisms of synbiotic in the
improvement of the epithelial barrier function, inflamma-
tory parameters (e.g., immune regulatory cell expansion, and
cytokines levels) and microbiota regulation, and the analysis
of SCFA still need to be further studied.
Notwithstanding these limitations, most importantly, our
results demonstrated that the synergistic synbiotic has par-
tially potentiated the intestinal mucosa from 5-FU-induced
intestinal mucositis compared to CIDCA 133 and FOS. Fur-
thermore, the primary mechanism used by the synbiotic to
ameliorate intestinal mucosa architecture included modula-
tion of inflammatory parameters, epithelial barrier markers,
and reduction in intestinal permeability, demonstrating to be
a promising therapeutic approach to be explored to alleviate
the intestinal inflammatory damage caused by chemotherapy.
Acknowledgements The authors would like to acknowledge the Pró-
Reitoria de Pesquisa - Universidade Federal de Minas Gerais, Con-
selho Nacional de Desenvolvimento Científico e Tecnológico (CNPq),
Coordenação de Aperfeiçoamento de Pessoal de Nível Superior
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
235 Page 12 of 14
(CAPES), and Fundação de Amparo à Pesquisa do Estado de Minas
Gerais (FAPEMIG) for their financial support and fellowships. We
would also like to acknowledge the CIDCA center (Center for Research
and Development in Food Cryotechnology) and Pablo F. Pérez of the
National University of La Plata, Argentine, for bacterial strain supply.
Author contributions Conceptualization: PM-A, LCLJ, MMD, and
VA; Methodology: FALB, VLB, GMC, GAB, TFS, NDC-R, KDV, ÊF;
Formal analysis and investigation: LMT, LCLJ; Writing-original draft
preparation: LMT, MFA, ASF, GMC, LCLJ; Writing-review and edit-
ing: JGL, SOAF, VNC, MMD, PM-A, AB, EF, FSM, and VA; Super-
vision: VA; Funding acquisition: VA. All authors read and approved
the final manuscript.
Funding This research was funded by Fundação de Amparo à Pesquisa
do Estado de Minas Gerais -FAPEMIG (grant numbers: RED-00132-
16 and APQ-00593-14), and Conselho Nacional de Desenvolvimento
Científico e Tecnológico - CNPq) (grant number: 312045/2020-4).
Data availability The current study’s data are available from the cor-
responding author upon reasonable request.
Declarations
Conflict of interest The authors declare no conflict of interest.
Ethical approval The study was conducted according to the guide-
lines of the Brazilian College of Animal Experimentation (COBEA)
and approved by the Local Animal Experimental Ethics Committee
(CEUA-UFMG) (Protocol n º 34/2021).
Consent for publication All authors consent to the publication of this
article.
References
Al-Khrashi LA, Badr AM, AL-Amin MA, Mahran YF (2022) Thymol
ameliorates 5-fluorouracil-induced intestinal mucositis: evidence
of down-regulatory effect on TGF-β/MAPK pathways through
NF-κB. J Biochem Mol Toxicol. https:// doi. org/ 10. 1002/ jbt. 22932
Andrade MER, Trindade LM, Leocádio PCL etal (2023) Associa-
tion of Fructo-oligosaccharides and arginine improves severity
of Mucositis and modulate the intestinal microbiota. Probiot-
ics Antimicrob Proteins 15:424–440. https:// doi. org/ 10. 1007/
s12602- 022- 10032-8
Barbosa MM, de Araújo AA, de Araújo Júnior RF etal (2018) Tel-
misartan modulates the oral mucositis induced by 5-fluorouracil in
hamsters. Front Physiol. https:// doi. org/ 10. 3389/ fphy s. 2018. 01204
Barroso FAL, de Jesus LCL, de Castro CP etal (2021) Intake of Lacto-
bacillus delbrueckii (pExu:hsp65) prevents the inflammation and
the disorganization of the intestinal mucosa in a mouse model of
Mucositis. Microorganisms 9:107. https:// doi. org/ 10. 3390/ micro
organ isms9 010107
Barroso FAL, de Jesus LCL, da Silva TF etal (2022) Lactobacil-
lus delbrueckii CIDCA 133 ameliorates chemotherapy-induced
Mucositis by modulating epithelial barrier and TLR2/4/Myd88/
NF-κB signaling pathway. Front Microbiol. https:// doi. org/ 10.
3389/ fmicb. 2022. 858036
Batista VL, da Silva TF, de Jesus LCL etal (2020) Probiotics, prebi-
otics, synbiotics, and paraprobiotics as a therapeutic alternative
for intestinal mucositis. Front Microbiol. https:// doi. org/ 10. 3389/
fmicb. 2020. 544490
Batista VL, De Jesus LCL, Tavares LM etal (2022) Paraprobiotics
and postbiotics of Lactobacillus delbrueckii CIDCA 133 mitigate
5-FU-induced intestinal inflammation. Microorganisms 10:1418.
https:// doi. org/ 10. 3390/ micro organ isms1 00714 18
Cao P, Wu L, Wu Z etal (2019) Effects of oligosaccharides on the
fermentation properties of Lactobacillus plantarum. J Dairy Sci
102:2863–2872. https:// doi. org/ 10. 3168/ jds. 2018- 15410
Capitán-Cañadas F, Ocón B, Aranda CJ etal (2016) Fructooligosac-
charides exert intestinal anti-inflammatory activity in the CD4 +
CD62L + T cell transfer model of colitis in C57BL/6J mice. Eur
J Nutr 55:1445–1454. https:// doi. org/ 10. 1007/ s00394- 015- 0962-6
Carvalho PLA, Andrade MER, Trindade LM etal (2021) Prophylactic
and therapeutic supplementation using fructo-oligosaccharide
improves the intestinal homeostasis after mucositis induced by
5- fluorouracil. Biomed Pharmacother 133:111012. https:// doi.
org/ 10. 1016/j. biopha. 2020. 111012
Carvalho R, Vaz A, Pereira FL etal (2018) Gut microbiome modu-
lation during treatment of mucositis with the dairy bacterium
Lactococcus lactis and recombinant strain secreting human
antimicrobial PAP. Sci Rep 8:15072. https:// doi. org/ 10. 1038/
s41598- 018- 33469-w
Chang C-W, Liu C-Y, Lee H-C etal (2018) Lactobacillus casei vari-
ety rhamnosus probiotic preventively attenuates 5-fluorouracil/
oxaliplatin-induced intestinal injury in a syngeneic colorectal
cancer model. Front Microbiol. https:// doi. org/ 10. 3389/ fmicb.
2018. 00983
Chang C-W, Lee H-C, Li L-H etal (2020) Fecal microbiota transplanta-
tion prevents intestinal Injury, upregulation of toll-like receptors,
and 5-fluorouracil/oxaliplatin-induced toxicity in colorectal can-
cer. Int J Mol Sci 21:386. https:// doi. org/ 10. 3390/ ijms2 10203 86
Coelho-Rocha ND, de Jesus LCL, Barroso FAL etal (2022) Evalua-
tion of probiotic properties of novel Brazilian Lactiplantibacillus
plantarum strains. Probiotics AntimicrobProteins. https:// doi. org/
10. 1007/ s12602- 022- 09978-6
Crombie J, Longo DL (2016) Principles of Cancer Treatment. Clinical
cardio-oncology. Elsevier, Amsterdam, pp 1–13
Dahlgren D, Sjöblom M, Hellström PM, Lennernäs H (2021) Chem-
otherapeutics-induced intestinal mucositis: pathophysiology and
potential treatment strategies. Front Pharmacol. https:// doi. org/
10. 3389/ fphar. 2021. 681417
Davani-Davari D, Negahdaripour M, Karimzadeh I etal (2019) Prebi-
otics: definition, types, sources, mechanisms, and clinical applica-
tions. Foods 8:92. https:// doi. org/ 10. 3390/ foods 80300 92
De Jesus LCL, Drumond MM, de Carvalho A etal (2019) Protective
effect of Lactobacillus delbrueckii subsp. Lactis CIDCA 133 in
a model of 5 fluorouracil-induced intestinal mucositis. J Funct
Foods 53:197–207. https:// doi. org/ 10. 1016/j. jff. 2018. 12. 027
de Jesus LCL, de Jesus Sousa T, Coelho-Rocha ND etal (2021) Safety
evaluation of Lactobacillus delbrueckii subsp.lactis CIDCA 133:
a health-promoting bacteria. Probiotics Antimicrob Proteins.
https:// doi. org/ 10. 1007/ s12602- 021- 09826-z
de Jesus LCL, Drumond MM, Aburjaile FF etal (2021) Probiog-
enomics of Lactobacillus delbrueckii subsp. lactis CIDCA 133:
in Silico, invitro, and invivo approaches. Microorganisms 9:829.
https:// doi. org/ 10. 3390/ micro organ isms9 040829
de Moraes ACF, da Silva IT, de Almeida-Pititto B, Ferreira SRG (2014)
Microbiota intestinal e risco cardiometabólico: mecanismos e
modulação dietética. Arquivos Brasileiros de Endocrinologia &
Metabologia 58:317–327. https:// doi. org/ 10. 1590/ 0004- 27300
00002 940
Dou Y, Yu X, Luo Y etal (2022) Effect of Fructooligosaccharides
supplementation on the gut microbiota in human: a systematic
review and meta-analysis. Nutrients 14:3298. https:// doi. org/ 10.
3390/ nu141 63298
Galdino FMP, Andrade MER, de Barros PAV etal (2018) Pretreatment
and treatment with fructo-oligosaccharides attenuate intestinal
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
Page 13 of 14 235
mucositis induced by 5-FU in mice. J Funct Foods 49:485–492.
https:// doi. org/ 10. 1016/j. jff. 2018. 09. 012
Gibson GR, Hutkins R, Sanders ME etal (2017) Expert consensus
document: the International Scientific Association for Probiot-
ics and Prebiotics (ISAPP) consensus statement on the defini-
tion and scope of prebiotics. Nat Reviews Gastroenterol Hepatol
14:491–502. https:// doi. org/ 10. 1038/ nrgas tro. 2017. 75
Giulietti A, Overbergh L, Valckx D etal (2001) An overview of
real-time quantitative PCR: applications to quantify cytokine
gene expression. Methods 25:386–401. https:// doi. org/ 10. 1006/
meth. 2001. 1261
Gomes-Santos AC, de Oliveira RP, Moreira TG etal (2017) Hsp65-
producing Lactococcus lactis prevents inflammatory intestinal
disease in mice by IL-10- and TLR2-dependent pathways. Front
Immunol. https:// doi. org/ 10. 3389/ fimmu. 2017. 00030
Hugo AA, Rolny IS, Romanin D, Pérez PF (2017) Lactobacil-
lus delbrueckii subsp. lactis (strain CIDCA 133) stimulates
murine macrophages infected with Citrobacter rodentium.
World J Microbiol Biotechnol 33:48. https:// doi. org/ 10. 1007/
s11274- 017- 2219-4
Ihara S, Hirata Y, Koike K (2017) TGF-β in inflammatory bowel dis-
ease: a key regulator of immune cells, epithelium, and the intes-
tinal microbiota. J Gastroenterol 52:777–787. https:// doi. org/ 10.
1007/ s00535- 017- 1350-1
Jung Y, Wen T, Mingler MK etal (2015) IL-1β in eosinophil-mediated
small intestinal homeostasis and IgA production. Mucosal Immu-
nol 8:930–942. https:// doi. org/ 10. 1038/ mi. 2014. 123
Kostler WJ, Hejna M, Wenzel C, Zielinski CC (2001) Oral mucositis
complicating chemotherapy and/or radiotherapy: options for pre-
vention and treatment. Cancer J Clin 51:290–315. https:// doi. org/
10. 3322/ canjc lin. 51.5. 290
Li H-L, Lu L, Wang X-S etal (2017) Alteration of gut microbiota
and inflammatory cytokine/chemokine profiles in 5-fluorouracil
induced intestinal mucositis. Front Cell Infect Microbiol. https://
doi. org/ 10. 3389/ fcimb. 2017. 00455
Liao M, Zhang Y, Qiu Y etal (2021) Fructooligosaccharide supple-
mentation alleviated the pathological immune response and pre-
vented the impairment of intestinal barrier in DSS-induced acute
colitis mice. Food Funct 12:9844–9854. https:// doi. org/ 10. 1039/
D1FO0 1147B
Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression
data using real-time quantitative PCR and the 2–∆∆CT method.
Methods 25:402–408. https:// doi. org/ 10. 1006/ meth. 2001. 1262
Longley DB, Harkin DP, Johnston PG (2003) 5-Fluorouracil: mecha-
nisms of action and clinical strategies. Nat Rev Cancer 3:330–338.
https:// doi. org/ 10. 1038/ nrc10 74
Mao B, Gu J, Li D etal (2018) Effects of different doses of Fructooli-
gosaccharides (FOS) on the composition of mice fecal microbiota,
especially the Bifidobacterium Composition. Nutrients 10:1105.
https:// doi. org/ 10. 3390/ nu100 81105
Markowiak P, Śliżewska K (2017) Effects of probiotics, prebiotics, and
synbiotics on human health. Nutrients 9:1021. https:// doi. org/ 10.
3390/ nu909 1021
Nakao T, Kurita N, Komatsu M etal (2012) Irinotecan injures tight
junction and causes bacterial translocation in rat. J Surg Res
173:341–347. https:// doi. org/ 10. 1016/j. jss. 2010. 10. 003
Parada Venegas D, De la Fuente MK, Landskron G etal (2019) Short
chain fatty acids (SCFAs)-mediated gut epithelial and immune
regulation and its relevance for inflammatory bowel diseases.
Front Immunol. https:// doi. org/ 10. 3389/ fimmu. 2019. 00277
Passalacqua KD, Charbonneau M-E, O’Riordan MXD (2016) Bacterial
metabolism shapes the host-pathogen interface. Microbiol Spectr.
https:// doi. org/ 10. 1128/ micro biols pec. VMBF- 0027- 2015
Pedroso SHSP, Vieira AT, Bastos RW etal (2015) Evaluation of
mucositis induced by irinotecan after microbial colonization in
germ-free mice. Microbiology 161:1950–1960. https:// doi. org/
10. 1099/ mic.0. 000149
Pourmasoumi M, Najafgholizadeh A, Hadi A etal (2019) The effect of
synbiotics in improving Helicobacter pylori eradication: a system-
atic review and meta-analysis. Complement Ther Med 43:36–43.
https:// doi. org/ 10. 1016/j. ctim. 2019. 01. 005
Quintanilha MF, Miranda VC, Souza RO etal (2022) Bifidobacterium
longum subsp. longum 51A attenuates intestinal injury against iri-
notecan-induced mucositis in mice. Life Sci 289:120243. https://
doi. org/ 10. 1016/j. lfs. 2021. 120243
Rolny IS, Tiscornia I, Racedo SM etal (2016) Lactobacillus del-
brueckii subsp lactis CIDCA 133 modulates response of human
epithelial and dendritic cells infected with Bacillus cereus. Benefi-
cial Microbes 7:749–760. https:// doi. org/ 10. 3920/ BM2015. 0191
Rooks MG, Garrett WS (2016) Gut microbiota, metabolites and host
immunity. Nat Rev Immunol 16:341–352. https:// doi. org/ 10. 1038/
nri. 2016. 42
Russo E, Giudici F, Fiorindi C etal (2019) Immunomodulating activ-
ity and therapeutic effects of short chain fatty acids and trypto-
phan post-biotics in inflammatory bowel disease. Front Immunol.
https:// doi. org/ 10. 3389/ fimmu. 2019. 02754
Savassi B, Cordeiro BF, Silva SH etal (2021) Lyophilized symbiotic
mitigates mucositis induced by 5-fluorouracil. Front Pharmacol.
https:// doi. org/ 10. 3389/ fphar. 2021. 755871
Shen S-R, Chen W-J, Chu H-F etal (2021) Amelioration of 5-fluoro-
uracil-induced intestinal mucositis by Streptococcus thermophilus
ST4 in a mouse model. PLoS ONE 16:e0253540. https:// doi. org/
10. 1371/ journ al. pone. 02535 40
Sheng K, He S, Sun M etal (2020) Synbiotic supplementation contain-
ing Bifidobacterium infantis and xylooligosaccharides alleviates
dextran sulfate sodium-induced ulcerative colitis. Food Funct
11:3964–3974. https:// doi. org/ 10. 1039/ D0FO0 0518E
Shida K, Nanno M, Nagata S (2011) Flexible cytokine production by
macrophages and T cells in response to probiotic bacteria: a possi-
ble mechanism by which probiotics exert multifunctional immune
regulatory activities. Gut Microbes 2:109–114. https:// doi. org/ 10.
4161/ gmic.2. 2. 15661
Smith CL, Geier MS, Yazbeck R etal (2008) Lactobacillus fermentum
BR11 and fructo-oligosaccharide partially reduce Jejunal inflam-
mation in a model of intestinal mucositis in rats. Nutr Cancer
60:757–767. https:// doi. org/ 10. 1080/ 01635 58080 21928 41
Soares PMG, Mota JMSC, Gomes AS etal (2008) Gastrointestinal
dysmotility in 5-fluorouracil-induced intestinal mucositis outlasts
inflammatory process resolution. Cancer Chemother Pharmacol
63:91–98. https:// doi. org/ 10. 1007/ s00280- 008- 0715-9
Soleimani A, Motamedzadeh A, Zarrati Mojarrad M etal (2019) The
effects of synbiotic supplementation on metabolic status in dia-
betic patients undergoing hemodialysis: a randomized, double-
blinded, placebo-controlled trial. Probiotics Antimicrob Proteins
11:1248–1256. https:// doi. org/ 10. 1007/ s12602- 018- 9499-3
Song M-K, Park M-Y, Sung M-K (2013) 5-Fluorouracil-induced
changes of intestinal integrity biomarkers in BALB/C mice. J Can-
cer Prev 18:322–329. https:// doi. org/ 10. 15430/ JCP. 2013. 18.4. 322
Sonis ST (2004) The pathobiology of mucositis. Nat Rev Cancer
4:277–284. https:// doi. org/ 10. 1038/ nrc13 18
Sougiannis AT, VanderVeen BN, Davis JM etal (2021) Understand-
ing chemotherapy-induced intestinal mucositis and strategies to
improve gut resilience. Am J Physiology-Gastrointestinal Liver
Physiol 320:G712–G719. https:// doi. org/ 10. 1152/ ajpgi. 00380.
2020
Souza DG, Cara DC, Cassali GD etal (2000) Effects of the PAF recep-
tor antagonist UK74505 on local and remote reperfusion injuries
following ischaemia of the superior mesenteric artery in the rat.
Br J Pharmacol 131:1800–1808. https:// doi. org/ 10. 1038/ sj. bjp.
07037 56
World Journal of Microbiology and Biotechnology (2023) 39:235
1 3
235 Page 14 of 14
Strath M, Warren DJ, Sanderson CJ (1985) Detection of eosinophils
using an eosinophil peroxidase assay. Its use as an assay for eosin-
ophil differentiation factors. J Immunol Methods 83:209–215.
https:// doi. org/ 10. 1016/ 0022- 1759(85) 90242-X
Swanson KS, Gibson GR, Hutkins R etal (2020) The International
Scientific Association for Probiotics and Prebiotics (ISAPP) con-
sensus statement on the definition and scope of synbiotics. Nat
Reviews Gastroenterol Hepatol 17:687–701. https:// doi. org/ 10.
1038/ s41575- 020- 0344-2
Touchefeu Y, Montassier E, Nieman K etal (2014) Systematic review:
the role of the gut microbiota in chemotherapy- or radiation-
induced gastrointestinal mucositis—Current evidence and poten-
tial clinical applications. Aliment Pharmacol Ther 40:409–421
Trindade LM, Martins VD, Rodrigues NM etal (2018) Oral adminis-
tration of Simbioflora® (synbiotic) attenuates intestinal damage
in a mouse model of 5-fluorouracil-induced mucositis. Beneficial
Microbes 9:477–486. https:// doi. org/ 10. 3920/ BM2017. 0082
van Vliet MJ, Harmsen HJM, de Bont ESJM, Tissing WJE (2010)
The role of intestinal microbiota in the development and severity
of chemotherapy-induced mucositis. PLoS Pathog 6:e1000879.
https:// doi. org/ 10. 1371/ journ al. ppat. 10008 79
Viana ML, Santos RGC, Generoso SV etal (2010) Pretreatment with
arginine preserves intestinal barrier integrity and reduces bacterial
translocation in mice. Nutrition 26:218–223. https:// doi. org/ 10.
1016/j. nut. 2009. 04. 005
Volynets V, Rings A, Bárdos G etal (2016) Intestinal barrier analy-
sis by assessment of mucins, tight junctions, and α-defensins
in healthy C57BL/6J and BALB/cJ mice. Tissue Barriers
4:e1208468. https:// doi. org/ 10. 1080/ 21688 370. 2016. 12084 68
Wardill HR, Bowen JM, Al-Dasooqi N etal (2014) Irinotecan disrupts
tight junction proteins within the gut. Cancer Biol Ther 15:236–
244. https:// doi. org/ 10. 4161/ cbt. 27222
Wei H-X, Wang B, Li B (2020) IL-10 and IL-22 in mucosal immunity:
driving protection and pathology. Front Immunol. https:// doi. org/
10. 3389/ fimmu. 2020. 01315
Wei L, Wen X-S, Xian CJ (2021) Chemotherapy-induced intestinal
microbiota dysbiosis impairs mucosal homeostasis by modulat-
ing toll-like receptor signaling pathways. Int J Mol Sci 22:9474.
https:// doi. org/ 10. 3390/ ijms2 21794 74
Wu J, Gan Y, Li M etal (2020) Patchouli alcohol attenuates 5-fluo-
rouracil-induced intestinal mucositis via TLR2/MyD88/NF-kB
pathway and regulation of microbiota. Biomed Pharmacother
124:109883. https:// doi. org/ 10. 1016/j. biopha. 2020. 109883
Wu W-JH, Kim M, Chang L-C etal (2022) Interleukin-1β secretion
induced by mucosa-associated gut commensal bacteria promotes
intestinal barrier repair. Gut Microbes. https:// doi. org/ 10. 1080/
19490 976. 2021. 20147 72
Xu X, Fukui H, Ran Y etal (2019) Alteration of GLP-1/GPR43
expression and gastrointestinal motility in dysbiotic mice treated
with vancomycin. Sci Rep 9:4381. https:// doi. org/ 10. 1038/
s41598- 019- 40978-9
Yeung C-Y, Chan W-T, Jiang C-B etal (2015) Amelioration of chemo-
therapy-induced intestinal mucositis by orally administered probi-
otics in a mouse model. PLoS ONE 10:e0138746. https:// doi. org/
10. 1371/ journ al. pone. 01387 46
Yeung C, Chiang Chiau J, Cheng M etal (2020) Modulations of probi-
otics on gut microbiota in a 5-fluorouracilinduced mouse model
of mucositis. J Gastroenterol Hepatol 35:806–814. https:// doi. org/
10. 1111/ jgh. 14890
Yue X, Wen S, Long-kun D etal (2022) Three important short-
chain fatty acids (SCFAs) attenuate the inflammatory response
induced by 5-FU and maintain the integrity of intestinal mucosal
tight junction. BMC Immunol 23:19. https:// doi. org/ 10. 1186/
s12865- 022- 00495-3
Zheng L, Zhang Y-L, Dai Y-C etal (2017) Jianpi Qingchang decoction
alleviates ulcerative colitis by inhibiting nuclear factor-κB activa-
tion. World J Gastroenterol 23:1180. https:// doi. org/ 10. 3748/ wjg.
v23. i7. 1180
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
Article
Lactobacillus delbrueckii subsp. lactis CIDCA 133 is a health-promoting bacterium that can alleviate gut inflammation and improve the epithelial barrier in a mouse model of mucositis. Despite these beneficial effects, the protective potential of this strain in other inflammation models, such as inflammatory bowel disease, remains unexplored. Herein, we examined for the first time the efficacy of Lactobacillus delbrueckii CIDCA 133 incorporated into a fermented milk formulation in the recovery of inflammation, epithelial damage, and restoration of gut microbiota in mice with dextran sulfate sodium-induced colitis. Oral administration of Lactobacillus delbrueckii CIDCA 133 fermented milk relieved colitis by decreasing levels of inflammatory factors (myeloperoxidase, N-acetyl-β-D-glucosaminidase, toll-like receptor 2, nuclear factor-κB, interleukins 10 and 6, and tumor necrosis factor), secretory immunoglobulin A levels, and intestinal paracellular permeability. This immunobiotic also modulated the expression of tight junction proteins (zonulin and occludin) and the activation of short-chain fatty acids-related receptors (G-protein coupled receptors 43 and 109A). Colonic protection was effectively associated with acetate production and restoration of gut microbiota composition. Treatment with Lactobacillus delbrueckii CIDCA 133 fermented milk increased the abundance of Firmicutes members (Lactobacillus genus) while decreasing the abundance of Proteobacteria (Helicobacter genus) and Bacteroidetes members (Bacteroides genus). These promising outcomes influenced the mice’s mucosal healing, colon length, body weight, and disease activity index, demonstrating that this immunobiotic could be explored as an alternative approach for managing inflammatory bowel disease.
Article
Full-text available
Mucositis is defined as inflammatory and ulcerative lesions along of the gastrointestinal tract that leads to the imbalance of the intestinal microbiota. The use of compounds with action on the integrity of the intestinal epithelium and their microbiota may be a beneficial alternative for the prevention and/or treatment of mucositis. So, the aim of this study was to evaluate the effectiveness of the association of fructo-oligosaccharides (FOS) and arginine on intestinal damage in experimental mucositis. BALB/c mice were randomized into five groups: CTL (without mucositis + saline), MUC (mucositis + saline), MUC + FOS (mucositis + supplementation with FOS—1st until 10th day), MUC + ARG (mucositis + supplementation with arginine—1st until 10th day), and MUC + FOS + ARG (mucositis + supplementation with FOS and arginine—1st until 10th day). On the 7th day, mucositis was induced with an intraperitoneal injection of 300 mg/kg 5-fluorouracil (5-FU), and after 72 h, the animals were euthanized. The results showed that association of FOS and arginine reduced weight loss and oxidative stress (P < 0.05) and maintained intestinal permeability and histological score at physiological levels. The supplementation with FOS and arginine also increased the number of goblet cells, collagen area, and GPR41 and GPR43 gene expression (P < 0.05). Besides these, the association of FOS and arginine modulated intestinal microbiota, leading to an increase in the abundance of the genera Bacteroides, Anaerostipes, and Lactobacillus (P < 0.05) in relation to increased concentration of propionate and acetate. In conclusion, the present results show that the association of FOS and arginine could be important adjuvants in the prevention of intestinal mucositis probably due to modulated intestinal microbiota.
Article
Full-text available
The human gut microbiota is highly heterogenous between individuals and also exhibits considerable day-to-day variation within individuals. We hypothesized that diet contributed to such inter- and/or intra-individual variance. Hence, we investigated the extent to which diet normalization impacted microbiota heterogeneity. We leveraged the control arm of our recently reported controlled-feeding study in which nine healthy individuals consumed a standardized additive-free diet for 10 days. Diet normalization did not impact inter-individual differences but reduced the extent of intra-individual day-to-day variation in fecal microbiota composition. Such decreased heterogeneity reflected individual-specific enrichment and depletion of an array of taxa microbiota members and was paralleled by a trend toward reduced intra-individual variance in fecal LPS and flagellin, which, collectively, reflect microbiota’s pro-inflammatory potential. Yet, the microbiota of some subjects did not change significantly over the course of the study, suggesting heterogeneity in microbiota resilience to dietary stress or that baseline diets of some subjects were perhaps similar to our study’s standardized diet. Collectively, our results indicate that short-term diet heterogeneity contributes to day-to-day intra-individual microbiota composition variance.
Article
Full-text available
Beneficial effects of Lactiplantibacillus plantarum strains have been widely reported. Knowing that the effects of probiotic bacteria are strain-dependent, this study aimed to characterize the probiotic properties and investigate the gastrointestinal protective effects of nine novel L. plantarum strains isolated from Bahia, Brazil. The probiotic functionality was first evaluated in vitro by characterizing bile salt and acidic tolerance, antibacterial activity, and adhesion to Caco-2 cells. Antibiotic resistance profile, mucin degradation, and hemolytic activity assays were also performed to evaluate safety features. In vivo analyses were conducted to investigate the anti-inflammatory effects of the strains on a mouse model of 5-Fluorouracil-induced mucositis. Our results suggest that the used L. plantarum strains have good tolerance to bile salts and low pH and can inhibit commonly gastrointestinal pathogens. Lp2 and Lpl1 strains also exhibited high adhesion rates to Caco-2 cells (13.64 and 9.05%, respectively). Phenotypical resistance to aminoglycosides, vancomycin, and tetracycline was observed for most strains. No strain showed hemolytic or mucolytic activity. Seven strains had a protective effect against histopathological and inflammatory damage induced by 5-FU. Gene expression analysis of inflammatory markers showed that five strains upregulated interleukin 10 (Il10), while four downregulated both interleukin 6 (Il6) and interleukin 1b (Il1b). Additionally, all strains reduced eosinophilic and neutrophilic infiltration; however, they could not prevent weight loss or reduced liquid/ food intake. Altogether, our study suggests these Brazilian L. plantarum strains present good probiotic characteristics and safety levels for future applications and can be therapeutically adjuvant alternatives to prevent/treat intestinal mucositis.
Article
Full-text available
Background: Numerous studies have investigated the effects of the supplementation of fructooligosaccharides (FOS) on the number of bacteria in the gut that are good for health, but the results have been inconsistent. Additionally, due to its high fermentability, supplementation of FOS may be associated with adverse gastrointestinal symptoms such as bloating and flatulence. Therefore, we assessed the effects of FOS interventions on the composition of gut microbiota and gastrointestinal symptoms in a systematic review and meta-analysis. Design: All randomized controlled trials published before 10 July 2022 that investigated the effects of FOS supplementation on the human gut microbiota composition and gastrointestinal symptoms and met the selection criteria were included in this study. Using fixed or random-effects models, the means and standard deviations of the differences between the two groups before and after the intervention were combined into weighted mean differences using 95% confidence intervals (CIs). Results: Eight studies containing 213 FOS supplements and 175 controls remained in this meta-analysis. Bifidobacterium spp. counts significantly increased during FOS ingestion (0.579, 95% CI: 0.444–0.714) in comparison with that of the control group. Subgroup analysis showed greater variation in Bifidobacterium spp. in adults (0.861, 95% CI: 0.614–1.108) than in infants (0.458, 95% CI: 0.297–0.619). The increase in Bifidobacterium spp. counts were greater in the group with an intervention duration greater than 4 weeks (0.841, 95% CI: 0.436–1.247) than an intervention time less than or equal to four weeks (0.532, 95% CI: 0.370–0.694), and in the group with intervention doses > 5 g (1.116, 95% CI: 0.685–1.546) the counts were higher than those with doses ≤ 5 g (0.521, 95% CI: 0.379–0.663). No differences in effect were found between FOS intervention and comparators in regard to the abundance of other prespecified bacteria or adverse gastrointestinal symptoms. Conclusions: This is the first meta-analysis to explore the effect of FOS on gut microbiota and to evaluate the adverse effects of FOS intake on the gastrointestinal tract. FOS supplementation could increase the number of colonic Bifidobacterium spp. while higher dose (7.5–15 g/d) and longer duration (>4 weeks) showed more distinct effects and was well tolerated.
Article
Full-text available
Intestinal mucositis is a commonly reported side effect in oncology practice. Probiotics are considered an excellent alternative therapeutic approach to this debilitating condition; however, there are safety questions regarding the viable consumption of probiotics in clinical practice due to the risks of systemic infections, especially in immune-compromised patients. The use of heat-killed or cell-free supernatants derived from probiotic strains has been evaluated to minimize these adverse effects. Thus, this work evaluated the anti-inflammatory properties of paraprobiotics (heat-killed) and postbiotics (cell-free supernatant) of the probiotic Lactobacillus delbrueckii CIDCA 133 strain in a mouse model of 5-Fluorouracil drug-induced mucositis. Administration of paraprobiotics and postbiotics reduced the neutrophil cells infiltrating into the small intestinal mucosa and ameliorated the intestinal epithelium architecture damaged by 5-FU. These ameliorative effects were associated with a downregulation of inflammatory markers (Tlr2, Nfkb1, Il12, Il17a, Il1b, Tnf), and upregulation of immunoregulatory Il10 cytokine and the epithelial barrier markers Ocln, Cldn1, 2, 5, Hp and Muc2. Thus, heat-killed L. delbrueckii CIDCA 133 and supernatants derived from this strain were shown to be effective in reducing 5-FU-induced inflammatory damage, demonstrating them to be an alternative approach to the problems arising from the use of live beneficial microorganisms in clinical practice.
Article
Full-text available
Intestinal mucositis promoted by the use of anticancer drugs is characterized by ulcerative inflammation of the intestinal mucosa, a debilitating side effect in cancer patients undergoing treatment. Probiotics are a potential therapeutic option to alleviate intestinal mucositis due to their effects on epithelial barrier integrity and anti-inflammatory modulation. This study investigated the health-promoting impact of Lactobacillus delbrueckii CIDCA 133 in modulating inflammatory and epithelial barrier markers to protect the intestinal mucosa from 5-fluorouracil-induced epithelial damage. L. delbrueckii CIDCA 133 consumption ameliorated small intestine shortening, inflammatory cell infiltration, intestinal permeability, villus atrophy, and goblet cell count, improving the intestinal mucosa architecture and its function in treated mice. Upregulation of Muc2, Cldn1, Hp, F11r, and Il10, and downregulation of markers involved in NF-κB signaling pathway activation (Tlr2, Tlr4, Nfkb1, Il6, and Il1b) were observed at the mRNA level. This work suggests a beneficial role of L. delbrueckii strain CIDCA 133 on intestinal damage induced by 5-FU chemotherapy through modulation of inflammatory pathways and improvement of epithelial barrier function.
Article
Full-text available
Background 5-Fluorouracil (5-FU) is a used chemotherapy drug for cancer, and its main side effect is intestinal mucositis which causes chemotherapy to fail. It was known that short-chain fatty acids (SCFAs) can inhibit immune cell release of various proinflammatory factors and inhibit excessive intestinal inflammation. However, the inhibitory effect of SCFAs on 5-FU-induced intestinal mucositis is still unclear. Results To simulate the effects of SCFAs on immune and intestinal epithelial cells, the cells (THP-1 cells and Caco-2 cells) were pretreated with sodium acetate (NaAc), sodium propionate (NaPc) and sodium butyrate (NaB), then inflammation was induced by 5-FU. The expressions of reactive oxygen species (ROS), Beclin-1, LC3-II, NF-κB p65, NLRP3 inflammasome, proinflammatory/anti-inflammatory cytokines and mucosal tight junction proteins were determined. In our results, the three SCFAs could inhibit ROS expressions, NLRP3, Caspase-1, IL-1β, IL-6, IL-18, Beclin-1 and LC3-II, when induced by 5-FU. In a 5-FU-induced chemoentermuctis mouse model, Lactobacillus rhamnoides can increase the concentrations of three SCFAs in faeces and increase the concentrations of IL-1β, IL-6 and IgA in serum, and decrease the expressions of NLRP3 and IL-17 in spleen cells. The expressions of ZO-1 and Occludin in intestinal mucosa were significantly increased. Conclusions These results indicated that the three SCFAs can effectively suppress the inflammation of THP-1 cells and Caco-2 cells and maintain tight junction integrity in intestinal mucosal epithelial cells.
Article
Full-text available
Mucositis is an adverse effect of cancer chemotherapies using 5-Fluorouracil (5-FU). It is characterized by mucosal inflammation, pain, diarrhea, and weight loss. Some studies reported promising healing effects of probiotic strains, when associated with prebiotics, as adjuvant treatment of mucositis. We developed a lyophilized symbiotic product, containing skimmed milk, supplemented with whey protein isolate (WPI) and with fructooligosaccharides (FOS), and fermented by Lactobacillus casei BL23, Lactiplantibacillus plantarum B7, and Lacticaseibacillus rhamnosus B1. In a mice 5-FU mucositis model, this symbiotic lyophilized formulation was able to reduce weight loss and intestinal permeability. This last was determined in vivo by quantifying blood radioactivity after oral administration of 99mTc-DTPA. Finally, histological damages caused by 5-FU-induced mucositis were monitored. Consumption of the symbiotic formulation caused a reduced score of inflammation in the duodenum, ileum, and colon. In addition, it decreased levels of pro-inflammatory cytokines IL-1β, IL-6, IL-17, and TNF-α in the mice ileum. The symbiotic product developed in this work thus represents a promising adjuvant treatment of mucositis.
Article
Full-text available
The gut microbiota is essential for maintenance and repair of the intestinal epithelial barrier. As shifts in both intestinal epithelial barrier function and microbiota composition are found in inflammatory bowel disease patients, it is critical to understand the role of distinct bacteria in regulating barrier repair. We identified a mouse commensal E. coli isolate, GDAR2-2, that protects mice from Citrobacter rodentium infection and dextran sulfate sodium-induced colitis. Colonization with GDAR2-2 in mice resulted in expansion of CX3CR1⁺ mononuclear phagocytes, including CX3CR1⁺ macrophages/dendritic cells and monocytes, along with IL-22-secreting type 3 innate lymphoid cells and improved epithelial barrier function. In vitro co-culture of macrophages with GDAR2-2 resulted in IL-1β production. In vivo, protection after GDAR2-2 colonization was lost after depletion of CX3CR1⁺ MNPs, or blockade of IL-1β or IL-22. We further identified human commensal E. coli isolates that similarly protect mice from C. rodentium infection through CX3CR1⁺ MNP and IL-1β production. Together, these findings demonstrate an unexpected role for commensal bacteria in promoting IL-1β secretion to support intestinal barrier repair.
Article
Intestinal mucositis (IM) is a critical side-effect associated with antineoplastic therapy. Treatment available is only palliative and often not effective. However, alternative therapeutic strategies, such as probiotics, have attracted significant attention due to their immune-modulatory action in several diseases. Thus, the present study aims to elucidate the therapeutic potential of the probiotic strain Bifidobacterium longum 51A in a murine model of mucositis induced by irinotecan. Due to the scarcity of studies on dose-response and viability (probiotic vs paraprobiotic), we first evaluated which dose and cell viability would be most effective in treating mucositis. In this study, the oral pretreatment with viable B. longum 51A at a concentration of 1 × 10⁹ CFU/mL reduced the daily disease activity index (p < 0.01), protected the intestinal architecture, preserved the length of the intestine (p < 0.05), and reduced intestinal permeability (p < 0.01), inflammation, and oxidative damage (p < 0.01) induced by irinotecan. Also, treatment with B. longum 51A increased the production of secretory immunoglobulin A (p < 0.05) in the intestinal fluid of mice with mucositis. Furthermore, B. longum 51A reversed the mucositis-induced increase in Enterobacteriaceae bacterial group in the gut (p < 0.01). In conclusion, these results showed that oral administration of B. longum 51A protects mice against intestinal damage caused by irinotecan, suggesting its use as a potential probiotic in therapy during mucositis.