You are on page 1of 18

Food &

Function
View Article Online
PAPER View Journal

Cooperative interactions between Veillonella ratti


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Cite this: DOI: 10.1039/d3fo03898j


and Lactobacillus acidophilus ameliorate
DSS-induced ulcerative colitis in mice†
Na Li,‡ Hejing Wang,‡ Huizhu Zhao, Mengyang Wang, Jin Cai, Yi Hao, Jia Yu,
Yun Jiang, Xin Lü and Bianfang Liu *

Veillonella and Lactobacillus species are key regulators of a healthy gut environment through metabolic
cross-feeding, influencing lactic acid and short-chain fatty acid (SCFA) levels, which are crucial for gut
health. This study aims to investigate how Veillonella ratti (V. ratti) and Lactobacillus acidophilus (LA) inter-
act with each other and alleviate dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in a mouse
model. We assess their metabolic interactions regarding carbon sources through co-culturing in a
modified medium. In the in vitro experiments, V. ratti and LA were inoculated in mono-cultures and co-
culture, and viable cell counts, OD600, pH, lactic acid, glucose and SCFAs were measured. For the in vivo
experiment, 60 C57BL/6 mice were randomly divided into five groups and administered V. ratti and LA
alone or in combination via oral gavage (1 × 109 CFU mL−1 per day per mouse) for 14 days. On the
seventh day, 2.5% DSS was added to the drinking water to induce colitis. The effects of these probiotics
on UC were evaluated by assessing intestinal barrier integrity and intestinal inflammation in the gut micro-
environment. In vitro results demonstrated that co-culturing V. ratti with LA significantly increased viable
cell numbers, lactic acid production, and SCFA production, while reducing pH and glucose levels in the
medium. In vivo findings revealed that intervention with V. ratti, particularly in combination with LA, alle-
viated symptoms, including weight loss, colon shortening, and tissue damage. These probiotics mitigated
intestinal inflammation by down-regulating pro-inflammatory molecules, such as IL-6, IL-1β, IL-γ, iNOS,
and IFN-γ, as well as oxidative stress markers, including MDA and MPO. Concurrently, they upregulated
the activity of anti-inflammatory enzymes, namely, SOD and GSH, and promoted the production of
SCFAs. The combined intervention of V. ratti and LA significantly increased acetic acid, propionic acid,
butyric acid, isobutyric acid, valeric acid, and total SCFAs in cecal contents. Furthermore, the intervention
Received 13th September 2023, of V. ratti and LA increased the abundance of beneficial bacteria, such as Akkermansia, while reducing the
Accepted 3rd October 2023
abundance of harmful bacteria, such as Escherichia–Shigella and Desulfovibrio, thereby mitigating exces-
DOI: 10.1039/d3fo03898j sive inflammation. These findings highlight the enhanced therapeutic effects resulting from the inter-
rsc.li/food-function actions between V. ratti and LA, demonstrating the potential of this combined probiotic approach.

1. Introduction lished that UC is associated with dysbiosis of intestinal flora,


disruption of mucosal barrier function, immune system altera-
Ulcerative colitis (UC) is a chronic inflammatory bowel disease tions, and a decrease in short-chain fatty acid (SCFA)-produ-
primarily affecting the mucosa and submucosa of the colon.1 cing bacteria and SCFAs.2 Managing UC has become a challen-
The DSS-induced UC mouse model exhibits oxidative damage ging task due to its high prevalence and complex pathogen-
and local inflammation, resulting in clinical symptoms such esis, the tendency to recur, and the potential development of
as diarrhea, weight loss, and colon lesions. It has been estab- colon cancer in some patients.3 Improving the gut mucosal
barrier integrity and the immune system, and adjusting the
gut microflora and their metabolites may contribute to redu-
College of Food Science and Engineering, Northwest A&F University, Yangling cing colonic inflammation.4 Given that current treatments for
712100, Shaanxi, China. E-mail: bfliu9509@163.com; Fax: +86-13519116643; UC are prone to cause severe undesirable side effects after
Tel: +86-13519116643
long-term use,5 therefore, there is an urgent need for novel,
† Electronic supplementary information (ESI) available. See DOI: https://doi.org/
10.1039/d3fo03898j safe, and effective preventive and therapeutic strategies for UC,
‡ These authors contributed equally to this work. such as probiotics.

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function

While the composition of the adult intestinal microbiota demonstrated that LUB significantly alleviated DSS-induced
exhibits interpersonal variation, certain core bacterial species colitis in mice, suggesting its potential as a probiotic for IBD
and their metabolites remain relatively stable.6 Among these treatment.22
core species, Lactobacillus spp. and Veillonella spp. have been Our previous research demonstrated that a high abundance
identified in the human intestinal flora. Their absence or low of Veillonella spp. in the intestines of piglets inhibited entero-
abundance has been associated with the manifestation of hemorrhagic Escherichia coli infection.23 Therefore, the present
symptoms.7 study aims to investigate the combined effects of Veillonella
Probiotics such as Lactobacillus plantarum, Bifidobacterium, ratti and Lactobacillus acidophilus on UC using an in vivo
Lactobacillus reuteri, and Lactobacillus acidophilus have been mouse model. The findings from this study may serve as a
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

utilized for the treatment and prevention of colitis.8–10 These basis for the development of potential probiotic combinations
lactic acid bacteria (LAB) produce substantial amounts of and have significant implications for future research on safe
lactic acid in the intestine. However, the accumulation of lactic and effective preventive and therapeutic strategies for UC.
acid is limited by lactate-utilizing bacteria (LUB) that metab-
olize it. The metabolic pathways of LUB are crucial in prevent-
ing excessive lactate accumulation and promoting intestinal 2. Materials and methods
ecosystem stability.11 Veillonella, an anaerobic commensal
LUB, plays a vital role in maintaining the stability and health 2.1 V. ratti and LA co-culture based on a modified
of the intestinal flora.6 It is essential for the establishment of 136 medium
healthy and mature gut anaerobic microbiota and the recovery 2.1.1 Preparation of the co-culture medium. V. ratti was
of the intestinal microbiota after antibiotic use.12 Additionally, isolated and identified in our laboratory and stored at the
Veillonella is also associated with the development of a healthy China Center of Industrial Culture Collection (CICC25149,
immune system in infants and promotes immune system GenBank accession no: OL597939.1). LA was obtained from
development in early childhood.13 Veillonella can metabolize Chr. Hansen China (CGMCC 21802).
lactic acid into acetic acid, propionic acid, and other SCFAs, To prepare the co-culture medium, we used a modified
which are vital for gut health. Intestinal SCFAs influence UC 136 medium based on the Veillonella medium base, as rec-
occurrence and development through multiple mechanisms, ommended by DSMZ (German Centre for Microbial Strain
including providing essential nutrients and energy to the host, Conservation, DSMZ136 medium). The ingredients of the
protecting the intestinal mucosal barrier, regulating intestinal medium per liter were as follows: trypticase (1 g), yeast extract
motility and host metabolism, and modulating the immune (0.6 g), Na-thioglycolate (0.75 g), Tween 80 (1 mL), glucose
system.2,14 Several studies have explored the physiological (8 g), putrescine (3 mg), (NH4)2SO4 (8 g), L-cysteine (0.5 g),
functions of Veillonella in maintaining the dynamic balance K2HPO4 (2 g), MgSO4·7H2O (0.5 g), MnSO4 (0.04 g), and di-
between lactic acid and SCFAs and its relevance to host health ammonium hydrogen citrate (2 g). The pH of the medium was
and disease.15,16 A few studies have observed lactic acid adjusted to 7.5 using K2CO3.
accumulation in the medium when Veillonella strains are co- 2.1.2 Detection of the OD, pH, and viable counts in the co-
cultured with Enterococcus faecium17 or found that the medium culture of V. ratti and LA. For the mono-cultures and co-
cultured with Lactobacillus and Veillonella isolated from culture experiments, 1.5% and 0.5% of the inoculums (OD600
chicken cecum inhibits pathogenic intestinal bacteria.18 = 0.5) of V. ratti and LA were respectively added into 10 mL of a
One of the crucial functions of Veillonella is the fermenta- modified 136 medium. The OD600 values and pH levels were
tion of organic acids to produce SCFAs; while some Veillonella measured using a spectrophotometer (UV spectrophotometer
strains cannot ferment carbohydrates such as glucose and 7200) and pH meter (PHSJ-3F), respectively. To determine the
lactose, they can ferment organic acids such as pyruvate and viable counts, the spread plate method was employed, where
lactic acid into acetic acid and propionic acid.19 On the other the samples were diluted and plated onto agar plates. The
hand, Lactobacillus acidophilus (LA) can ferment common plates were then incubated under suitable conditions, and the
sugars and plant-derived carbohydrates such as cellobiose and number of viable bacteria was calculated using colony forming
amygdalin.20 Some studies have demonstrated that cross- units. Samples from the mono-cultures and co-culture were
feeding interactions in co-culture can promote the growth of preserved at −80 °C for subsequent analysis of lactic acid,
the two bacteria by converting intermediates such as succi- glucose, and short-chain fatty acids (SCFAs).
nate, lactate, and acetyl CoA into acetate, propionate, butyrate, 2.1.3 Lactic acid and glucose assessment. To measure
and other SCFAs.21 lactate production and glucose uptake in the medium, a
Intestinal diseases, including UC and colorectal cancer, are lactate assay kit and a glucose assay kit (Jiancheng
associated with a decrease in the number of LUB. Patients Bioengineering Institute, Nanjing, China) were used. A colori-
with inflammatory bowel disease (IBD), unlike healthy individ- metric detection method was followed according to the manu-
uals, often exhibit high lactate accumulation in the colon, facturer’s instructions.
because it cannot be efficiently metabolized by LUB into host- 2.1.4 Detection of SCFAs in the co-culture of V. ratti and
friendly SCFAs.21 LUB plays a crucial role in maintaining the LA. For SCFA detection, the same method as described in
stability of the human colonic microbial ecosystem. Chen et al. section 2.9 was employed, with slight modifications in the

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper

sample preparation. Briefly, a bacterial culture (6 mL) was cen- diversity analysis. On the 22nd day, all mice were anesthetized
trifuged at 5000 rpm (4 °C) for 10 min. Next, 5 mL of the fil- and sacrificed. The entire colon tissues were harvested,
trate and 1 mL of 50% H2SO4 (v/v) solution were added to a excised, and rinsed with cold PBS for macroscopic assessment
centrifuge tube, followed by vortexing for 5 min. Subsequently, of changes.
5 mL of ether was added, and the mixture was allowed to
stand at 4 °C for 30 min, with intermittent shaking every 2.4. Disease activity indexes
5 min. Afterward, the solution was centrifuged at 5000 rpm After oral administration, daily measurements were taken of
(4 °C) for 20 min. Finally, the supernatant was extracted, con- the body weight, stool consistency, and bleeding of the mice.
centrated using nitrogen blowing, and redissolved in 1 mL of The severity of colitis was assessed using the disease activity
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

ether. index (DAI) score, as described by Mennigen et al.24 The DAI


score takes into account weight loss, stool consistency, and
2.2. Bacterial preparation for animal experiments bleeding as indicators of colitis severity. The presence of fecal
For the cultivation of V. ratti, BHI medium supplemented with occult blood was tested daily using a fecal occult blood kit
0.5% sodium lactate was used. The culture was incubated obtained from Nanjing Jiancheng Institute of Biological
under anaerobic conditions at 37 °C for 48 h. LA was cultured Engineering. Specific DAI scores were calculated using a pre-
in MRS broth at 37 °C for 12 h. viously described method (Table S1†).25
To collect the bacterial cells, the bacterial solution (10 mL)
was centrifuged at 8000 rpm (4 °C) for 10 minutes. The cells were 2.5. Histopathological evaluation
then washed twice with phosphate-buffered saline (PBS), resus- Approximately 1 cm sections from the proximal and distal
pended in saline, and adjusted to a concentration of 109 CFU colons of the mice were collected and fixed in a 4% (w/v) paraf-
mL−1 for subsequent administration via mouse gavage. It is ormaldehyde solution at 4 °C overnight. After fixation, the
important to note that the V. ratti suspension should be used colons were embedded in paraffin and sectioned into 5 μm
immediately, and the gavage procedure should be completed slices for hematoxylin and eosin (H&E) staining. The stained
within 1 hour of preparation due to its strict anaerobic nature. sections were observed and their images were captured using a
microscope (DP27 digital microscope, Olympus, Japan). The
2.3. Animal experiment assessment of the colonic mucosa damage score was per-
Sixty male C57BL/6 mice (SPF grade) weighing 20 ± 2 g and formed following a previously described method (Table S2†).26
aged 6–8 weeks were obtained from Hunan SJA Laboratory Co.,
Ltd (Hunan, China). The mice were housed under controlled 2.6. Real-time quantitative fluorescence polymerase chain
conditions at a temperature of 25 °C with a 12-hour light/dark reaction (RT-qPCR)
cycle. They had access to standard chow and sterilized water Total RNA was extracted from 50 mg of colon tissue using a
ad libitum. All animal experiments were conducted in compli- TaKaRa RNAiso Plus kit (Beijing Haiding, China) following the
ance with the guidelines and regulations approved by the manufacturer’s protocol. The extracted RNA was then qualitat-
Animal Ethics and Welfare Committee of Northwest A&F ively and quantitatively evaluated using a micro-spectrophoto-
University, China (SP2022015). meter Nano-200 (Hangzhou Allsheng Instruments, Korea).
The detailed experimental scheme is presented in Fig. 3A. Reverse transcription was performed using a first-strand cDNA
Following a week of acclimatization, the mice were randomly synthesis kit (Beijing Dining Biotechnology Co., Ltd), followed
assigned to five groups (n = 12): the control group with normal by RT-qPCR amplification of 2 μL dilutions of cDNA using a
drinking water (Ctrl), the DSS colitis group (DSS), the LA (109 CFX96 Touch™ real-time PCR detection system (Bio-Rad,
CFU mL−1, 200 μL per day) intervention group (DSS + LA), the Hercules, CA, USA) and PerfectStart® Green qPCR SuperMix
V. ratti (109 CFU mL−1, 200 μL per day) intervention group (DSS + (TransGen Biotech, Beijing, China). The primer sequences for
V. ratti), and the V. ratti (109 CFU mL−1, 100 μL per day) + LA (109 the target genes can be found in Table S3.† GAPDH house-
CFU mL−1, 100 μL per day) intervention group (DSS + mixture). keeping genes were used as an internal reference. The relative
During the first week of the acclimatization period, all expression of mRNA was calculated using the 2−ΔΔCt method.
groups were fed standard chow and purified water. In the
intervention period, the Ctrl and DSS groups received 200 μL 2.7. Detection of oxidative factors in the colon
of saline solution orally, while the probiotic intervention The levels of myeloperoxidase (MPO), malondialdehyde
groups received daily oral administration of 200 μL of bacterial (MDA), superoxide dismutase (SOD), and glutathione (GSH) in
suspension (109 CFU mL−1). mouse colonic tissues were measured using commercial kits
After the seven-day intervention period, the UC mouse (Jiancheng Bioengineering Institute, Nanjing, China) following
model was induced by daily administration of 2.5% DSS (w/v, the manufacturer’s instructions. To perform the measure-
MW: 36 000–50 000, American MP Biomedical Company) in the ments, 100 mg of colon tissue was homogenized with 0.9 mL
drinking water, while the bacterial interventions continued. of PBS buffer using a freezing grinder. The homogenate was
Throughout the experiment, the body weight, food intake, and then centrifuged at 10 000 rpm for 15 minutes at 4 °C, and the
water consumption of each mouse were recorded daily. On the resulting supernatant was collected. The protein concentration
14th and 21st days, fecal samples were collected for microbial in the supernatant was determined using a bicinchoninic acid

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function

(BCA) protein assay kit (Jiancheng Bioengineering Institute, experiments were presented as mean ± standard deviation
Nanjing, China). The activities of GSH, MDA, and SOD in the (SD). Statistical analysis of differences between multiple
colon were expressed as nanomoles per milligram of protein groups was conducted using two-way ANOVA, followed by
(nmol mg−1 protein) and units per milligram of protein (U Tukey’s multiple comparison test, with a significance level set
mg−1 protein). at P < 0.05.

2.8. Fecal microbiota analysis


Fecal samples from the mice were collected and immediately 3. Results
frozen at −80 °C. The frozen samples were then placed on dry
3.1. Cross-feeding between V. ratti and LA in a modified
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

ice and sent to LC Biotechnologies Co., Ltd (Hangzhou, China)


136 medium
for microbiota diversity analysis. Total DNA was extracted from
the fecal samples using an OMEGA stool DNA kit. The bac- In our previous experimental study, both V. ratti and LA
terial 16S rRNA genes in the V3–V4 region were amplified showed normal growth in the 136 medium (to be published).
using the forward primer 341F (5′- Considering the dynamic metabolic responses to changes in
CCTACGGGNGGCWGCAG-3′) and the reverse primer 805R (5′- carbon sources during the co-culture of these two strains, we
GACTACHVGGGTATCTAATCC-3′). The resulting amplicons adjusted the carbon source amount in the co-culture medium
were pooled in equal amounts, and paired-end sequencing accordingly. As depicted in Fig. 1, compared to mono-cultures,
with a read length of 2 × 250 bp was performed using the V. ratti co-cultured with LA exhibited an accelerated entry into
Illumina platform. After sequencing, all sequences were classi- the logarithmic growth phase, with a higher OD600 observed
fied into operational taxonomic units (OTUs) with a similarity from 6 hours and a significantly increased number of viable
level of 97%. To investigate the bacterial members responsible bacteria from 9 hours (P < 0.05, Fig. 1A and C). Additionally,
for the observed differences the between groups, LEfSe ana- the co-culture led to a noticeable decrease in the medium’s pH
lysis was conducted. compared to the mono-cultures (Fig. 1B). Measurements of
lactic acid and glucose in the medium demonstrated that the
2.9. Short-chain fatty acid (SCFA) and lactic acid detection co-culture of V. ratti with LA consumed more glucose and
Short-chain fatty acids (SCFAs) in the feces of the mice were exhibited enhanced lactic acid production compared to mono-
measured using a gas chromatograph (GC-2014C, Shimadzu cultures (P < 0.05, Fig. 1D and E).
Corporation, Japan) equipped with a DB-FFAP capillary Gas chromatography analysis was performed to detect the
column (30 m × 0.25 µm × 0.25 µm, Agilent Technologies, Palo short-chain fatty acids (SCFAs) produced in the co-culture and
Alto, CA, USA) and a flame ionization detector. Fecal samples mono-cultures of V. ratti and LA. The results showed that the
weighing 200 mg were dissolved, homogenized, and then cen- two bacteria significantly produced higher amounts of acetic
trifuged at 10 000 rpm (4 °C) for 10 minutes. The resulting acid (378.29 ± 4.34 mmol L−1), propionic acid (253.32 ±
supernatant was collected and filtered through a 0.22 µm 0.9 mmol L−1), isobutyric acid (162.35 ± 0.42 mmol L−1) and
organic membrane into gas-phase vials for GC-MS analysis. butyric acid (149.24 ± 0.49 mmol L−1), and total SCFAs
The gas chromatography-mass spectrometry (GC-MS) con- (1218.76 ± 3.92 mmol L−1) after 48 hours of co-culture (Fig. 2).
ditions were as follows: nitrogen (N2) was used as the carrier There was no significant difference in the production of valeric
gas with a split ratio of 10 : 1. Temperature programmed gas acid and isovaleric acid between the mono-cultures and the co-
chromatography (TPGC) involved an initial temperature of culture (data not shown). These findings indicate that the co-
50 °C for 1 minute, followed by an increase to 120 °C at a rate culture of V. ratti and LA promotes bacterial growth and pro-
of 15 °C min−1. The temperature was then raised to 170 °C at a liferation through cross-feeding interactions, where LA con-
rate of 5 °C min−1, and finally raised to 240 °C at 15 °C min−1 sumes glucose and converts it into lactic acid, which in turn
and held for 3 minutes. The injector and detector tempera- serves as a carbon source for V. ratti to produce SCFAs.
tures were set to 250 °C and 270 °C, respectively. Standard
SCFA curves were prepared using acetic acid, propionic acid, 3.2. V. ratti and LA restore body weight and relieve colitis
isobutyric acid, butyric acid, isovaleric acid, and valeric acid symptoms in DSS-induced colitis mice
(Sigma-Aldrich, St Louis, MO, USA). The effects of V. ratti and LA on colitis symptoms induced by DSS
Lactic acid in the feces of the mice was measured using a in mice were investigated (Fig. 3). The probiotic intervention was
lactate assay kit (Jiancheng Bioengineering Institute, Nanjing, assessed by monitoring the body weight, food intake, and general
China) through colorimetric detection following the manufac- conditions of the animals (Table S4†). Throughout the interven-
turer’s instructions. The supernatant solution of the feces of tion period, mice treated with V. ratti and LA for one week
the mice was standardized based on the sample weight. showed no signs of toxicity compared to the Ctrl group. All
groups of mice experienced weight loss during the UC model
2.10. Statistical analysis induction period (Fig. 3B). Notably, the DSS + mixture group
The trial data were analyzed and graphed for significance exhibited significantly less weight loss (P < 0.05).
using GraphPad Prism 8.0.1 software (GraphPad Software, Inc., Compared to the Ctrl group, the DAI score of the DSS group
San Diego, CA, USA). The data obtained from five independent began to increase on day three (P < 0.01, Fig. 3C). In contrast,

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 1 V. ratti and LA mono-cultures and co-culture in vitro. (a) The growth curves of the mono-culture and co-culture of V. ratti and LA; (b)
changes of pH in the mono-cultures and co-culture of V. ratti and LA; (c) changes in the number of viable cells in the mono-cultures and co-culture
of V. ratti and LA; (d) changes in the content of glucose; and (e) changes in the content of lactic acid; the analysis was performed in triplicate (n = 3).
All significant differences were analyzed using two-way ANOVA and Tukey’s post hoc test. LA compared with co-culture, *p < 0.05; **p < 0.01; ***p
< 0.001; ****p < 0.0001. V. ratti compared with co-culture, #p < 0.05; ##p < 0.01; ##p < 0.001; ####p < 0.0001.

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 2 The SCFAs produced in the co-culture and mono-cultures of the V. ratti and LA. (a)–(e) show acetic acid, propionic acid, butyric acid, isobu-
tyric acid, and the total short-chain fatty acids, respectively. The analysis was performed in triplicate (n = 3). All significant differences were analyzed
using two-way ANOVA and Tukey’s post hoc test. LA compared with co-culture, *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. V. ratti compared
with co-culture, #p < 0.05; ##p < 0.01; ##p < 0.001; ####p < 0.0001.

the DSS + V. ratti, DSS + LA, and DSS + mixture groups dis- Enlarged spleens and temporary shrinkage of the thymus
played significantly lower DAI scores than the DSS group on were observed in mice.
day seven (P < 0.01, P < 0.05, or P < 0.001). Fecal occult blood The weighing of the spleens showed that the thymus index
yielded showed negative results in the Ctrl group, with virtually exhibited a significant decrease, while the spleen index
no discoloration within 2 minutes, and positive results were showed a significant increase in the DSS group compared to
observed in the DSS group, with an immediate shift to dark the Ctrl group. Furthermore, the thymus index exhibited a sig-
blue-black. However, the levels of occult blood were minimal nificant decrease, while the spleen index showed a significant
in the DSS + V. ratti and DSS + mixture groups, indicating that increase in the DSS group compared to the Ctrl group.
V. ratti intake improved hematochezia in the mice (Fig. 3D). Importantly, the thymus index for the DSS + mixture group

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 3 V. ratti and LA ameliorate colitis clinical symptoms. (a) Experimental design and grouping schematic diagram. Colitis was induced by the
administration of 2.5% DSS in drinking water for 14 days. LA, V. ratti, or LA + V. ratti were supplemented in the saline solution for 7 days. Ctrl: healthy
control group fed with regular diet. (b) Rate of weight loss in mice during the UC model induction period; (c) schematic diagram of the trend of the
DAI score; (d) occult blood condition in the modeling period; (e) spleen index (%); and (f ) thymus index (%). All significant differences were analyzed
using one-way ANOVA and Tukey’s post hoc test. Compared with the Ctrl group, *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. Compared with
the DSS group, #p < 0.05; ##p < 0.01; ##p < 0.001; ####p < 0.0001.

and the Ctrl group showed no significant difference, F). These findings suggest that interventions of V. ratti and LA
suggesting that the intervention of V. ratti and LA led to partial promote body weight recovery and alleviate colitis symptoms
recovery of the health of the spleen and thymus (Fig. 3E and in DSS-induced colitis mice.

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function

3.3. V. ratti and LA reduce damage to the colon tissues 0.001), indicating damage to the tight junction structure. In
The colon length of mice (5.414 ± 0.2424 cm) was significantly contrast, the expression of occludin was significantly increased
shortened due to the DSS treatment compared to the Ctrl in the DSS + V. ratti group (P < 0.0001). These results indicate
group (8.786 ± 0.2807 cm) (Fig. 4A and B). However, the DSS + that the combination of V. ratti and LA may prevent DSS-
V. ratti, DSS + LA, and DSS + mixture intervention groups effec- induced disruption of intestinal integrity by effectively regulat-
tively prevented colon shortening compared to the DSS group ing the expression of related genes in the colon, including the
(6.786 ± 0.4562 cm, 6.443 ± 0.2010 cm, or 7.257 ± 0.2553 cm, enhanced mucin mRNA and tight junction protein expressions
respectively). (Fig. 5I).
In the Ctrl group, the mucosal epithelium appeared intact 3.5. V. ratti and LA alleviate oxidative stress in the colons of
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

and continuous, the mucosal villous glands were neatly the UC mice
arranged, the presence of lymphocytes in the lamina propria
was minimal, and there was no infiltration of inflammatory We assessed the levels of superoxide dismutase (SOD), malon-
cells. Conversely, the DSS group exhibited severe damage to dialdehyde (MDA), myeloperoxidase (MPO), and reduced gluta-
the colon tissues, including deformed crypt structures, thione (GSH) in the colonic homogenate supernatant of mice
destruction or even absence of goblet cells, extensive necrosis with colitis using a commercial kit. As shown in Fig. 6C and D,
of the colonic mucosal layer, damage to the mucosal epi- the levels of MDA and MPO were significantly elevated by
thelium, and infiltration of inflammatory cells. In contrast, approximately three-fold (P < 0.001) in the DSS group com-
both the DSS + V. ratti and DSS + mixture intervention groups pared to the Ctrl group. In contrast, the activity of SOD and the
effectively alleviated colitis. The mucosal tissues exhibited stra- levels of GSH were significantly decreased (P < 0.001; Fig. 6A
tification, the arrangement of mucosal villous glands was less and B) in the DSS group compared to the Ctrl group. However,
disordered, and the infiltration of inflammatory cells was sig- in the DSS + mixture group, the contents of SOD and GSH
nificantly reduced. Additionally, the DSS + mixture group were significantly increased (P < 0.01), while the levels of MDA
showed significantly lower histological scores and more intact and MPO were decreased compared to the DSS and individual
colonic tissue compared to the DSS + V. ratti group (P < 0.01; treatment groups (Fig. 6). These results suggest that the combi-
Fig. 4C and D). nation treatment of V. ratti and LA is more effective in alleviat-
ing oxidative stress of UC than the single treatment.

3.4. V. ratti and LA regulate the expression of related genes 3.6. V. ratti and LA contribute to restoring the intestinal flora
in the colon homeostasis of UC mice
To investigate whether V. ratti and LA regulated the expression We collected fecal samples from mice to analyze the compo-
of related genes in the colon, we conducted an assessment of sition of the gut microbiota. In total, we identified 31 genera
the mRNA expressions of TJ proteins occludin, mucins (MUC2 in all samples during the probiotic intervention period.
and MUC3) and cytokines (IL-6, IL-1β, IL-17a, IL-γ, IFN-γ, TNF- Importantly, the relative abundance of Ligilactobacillus and
α, and iNOS) in the colon tissue using RT-qPCR. The levels of total lactic acid bacteria (LAB) was significantly higher in the
inflammatory cytokines were significantly elevated in the DSS mixture group compared to the other groups (Fig. 7A–C, P <
group compared to the Ctrl group (P < 0.05; Fig. 5). However, 0.05), indicating that the interactions between V. ratti and LA
these inflammatory cytokines decreased significantly in the promotes the growth of LAB, which is consistent with the
DSS + V. ratti and DSS + mixture groups compared to the DSS in vitro results of this study.
group (P < 0.05). Notably, the DSS + mixture group exhibited a During the UC model induction period, DSS treatment sig-
pronounced effect in suppressing these inflammatory cyto- nificantly reduced the diversity of the gut microbiota (P < 0.05,
kines (P < 0.05). The level of TNF-α did not differ significantly Fig. 8A–E). Fig. 8F illustrates the 15 major phyla, with
between the groups (Fig. 5E). Additionally, the expression of Bacteroidetes and Firmicutes being the dominant ones. DSS
PPAR-γ was reduced in the DSS group (0.019 ± 0.0045; Fig. 5F) treatment increased the relative abundance of Firmicutes,
compared to the Ctrl group (1 ± 0.196). However, adminis- while decreasing the relative abundance of Bacteroidetes com-
tration of V. ratti and LA restored PPAR-γ expression levels in pared to the Ctrl group (P < 0.05, P < 0.01). This shift resulted
the colon of the mice in the mixture group (0.322 ± 0.039). in an elevated Firmicutes to Bacteroidetes ratio in the DSS
To further assess the preventative effect of V. ratti and LA group compared to the Ctrl group (P < 0.001, Fig. 8J). In con-
on the epithelial mucosa in the mice with DSS-induced colitis, trast, the F/B ratio was decreased in the DSS + mixture group
we examined the expression of MUC2 and MUC3 genes, which compared to the DSS group (P < 0.01). It is worth noting that
are crucial for mucosal integrity The expression of both genes the ratio of F/B has been reported to increase in the animal
was significantly down-regulated in the DSS group compared models of UC, which is consistent with our findings, However,
to the Ctrl group (P < 0.0001, P < 0.0001; Fig. 5I). However, the a combined intervention of V. ratti and LA significantly miti-
expression of these genes was prominently up-regulated in the gated this increase, indicating a positive impact on the gut
DSS + mixture group (P < 0.001, P < 0.1). Furthermore, the microbiota composition.
expression of the tight junction protein, occludin, was sup- Furthermore, we identified 30 genera in all samples
pressed in the DSS group compared to the Ctrl group (P < (Fig. 8G). The relative abundance of

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 4 V. ratti and LA interventions and the histopathological evaluations of their effects in the colon. (a) Macroscopic pictures of the colons; (b)
colon length of the mice; (c) histopathological score of the colon; and (d) H&E-stained sections of the mouse colon tissue (×10 and ×40). All signifi-
cant differences were analyzed using one-way ANOVA and Tukey’s post hoc test. Compared with the Ctrl group, *p < 0.05; **p < 0.01; ***p < 0.001;
****p < 0.0001. Compared with the DSS group, #p < 0.05; ##p < 0.01; ##p < 0.001; ####p < 0.0001.

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 5 V. ratti and LA regulated the expression of related genes in the colon. (a) Gene expression of IL-6 in the colonic epithelial tissue; (b) gene
expression of IL-1β in the colonic epithelial tissue; (c) gene expression of IL-17a in the colonic epithelial tissue; (d) gene expression of IL-γ in the
colonic epithelial tissue; (e) gene expression of TNF-α in the colonic epithelial tissue; (f ) gene expression of PPAR-γ in the colonic epithelial tissue;
(g) gene expression of IFN-γ in the colonic epithelial tissue; (h) gene expression of iNOS in the colonic epithelial tissue; and (i) gene expression of
tight junction proteins and mucins in the colonic epithelial tissue. All significant difference analyses were performed using one-way ANOVA and
Tukey’s post hoc test. Compared with the Ctrl group, *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. Compared with the DSS group, #p < 0.05;
##
p < 0.01; ###p < 0.001.

Desulfovibrionaceae_unclassified, Escherichia–Shigella, and Bacteroides were significantly increased in the DSS + mixture
Parasutterella was significantly increased, whereas the relative group compared to the DSS group, which indicates that the
abundance of Muribaculaceae_unclassified, Alloprevotella, intervention of V. ratti and LA enriched the two genera in the
Ruminococcus, Roseburia, and Ligilactobacillus was significantly intestines of mice.
decreased in the DSS group compared to the Ctrl group (P < To further analyze the gut microbiota diversity among the
0.05). However, the relative abundances of Akkermansia and five groups, we employed the LDA effect size (LEfSe) approach.

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 6 V. ratti and LA regulated oxidative stress in the colons of UC mice. (a) GSH; (b) SOD; (c) MDA; and (d) MPO. All significant difference analyses
were performed using one-way ANOVA and Tukey’s post hoc test. Compared with the Ctrl group, *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Compared with the DSS group, #p < 0.05; ##p < 0.01; ##p < 0.001; ####p < 0.0001.

The LDA score histogram was generated to identify statistically mixture group exhibited significantly increased levels of acetic
significant biomarkers and the dominant microorganisms in acid and isobutyric acid (P < 0.001), and reduced lactate
each group (Fig. 8H and I). Dominant communities were content (P < 0.0001) compared to the DSS, DSS + LA and DSS +
identified in the Ctrl, DSS, DSS + V. ratti, DSS + LA, and DSS + V. ratti groups. Importantly, the DSS + mixture group demon-
mixture groups, respectively. The Ctrl group showed higher strated a higher total SCFA content and a lower lactate content
relative abundances of the bacterial family Ruminococcaceae than the other groups (Fig. 9, P < 0.0001). These results indi-
and the genera Alloprevotella, Roseburia, and Muribaculum. In cate that when the UC mice were administered with a combi-
contrast, the DSS group exhibited a significant enrichment of nation of V. ratti and LA, the lactate produced by LA was effec-
potentially pathogenic bacteria, such as Proteobacteria, tively converted into SCFAs by V. ratti, thus maintaining intesti-
Desulfobacterota, Enterobacteriaceae, and Escherichia–Shigella. nal health and alleviating colitis in mice.
Notably, the DSS + mixture group showed an increase in ben-
eficial bacteria, including Verrucomicrobiota, Akkermansia and
Bacteroides (Fig. 8J).
4. Discussion
3.7. V. ratti and LA modulate the production of SCFAs, SCFA- Gut microbiota dysbiosis is a significant factor in the occurrence
producing bacteria and lactate and development of UC.27 Furthermore, patients with severe UC
We conducted an analysis of short-chain fatty acids (SCFAs) often exhibit elevated levels of fecal lactate.28 Previous studies
and lactate content in feces using gas chromatography (GC) have illuminated the beneficial impact of introducing feces-
and a lactate assay kit, respectively. The DSS group showed sig- enriched LUB flora in ameliorating colitis in murine models.22
nificantly reduced levels of acetic acid (P < 0.001), isobutyric Notably, L. acidophilus (LA) has shown promising potential in alle-
acid (P < 0.01), valeric acid (P < 0.05), isovaleric acid (P < 0.05), viating UC in mice by modulating immune responses, influen-
and total SCFAs, while lactate content was increased (P < cing miRNA expression, restoring gut microbiota balance, and
0.0001) compared to the Ctrl group. In contrast, the DSS + producing essential metabolites.10

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 7 Effect of V. ratti and LA pretreatment on the intestinal microbiota in healthy mice. During the intervention period: (a) the relative abundances
of the gut microbiota at the genus level; (b) the relative abundance of Ligilactobacillus; and (c) the total relative abundance of LAB. Compared with
the Ctrl group, **p < 0.01; compared with the DSS group, ##p < 0.01; DSS + V. ratti vs. DSS + mixture, *p < 0.05.

Veillonella, a key member of the LUB group found in the use of lactic acid bacteria as intestinal probiotics for
the intestines, actively engages in cross-feeding interactions UC, there remains a paucity of reports on the combined
with lactic acid bacteria to regulate the intestinal environ- probiotic effect of Veillonella and LA in relieving colitis in
ment of the host. While numerous studies have examined mice. Thus, in this study, we aimed to assess the probiotic

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 8 V. ratti and LA contributed to restore the intestinal flora homeostasis of UC mice. During the UC model induction period: (a) rarefaction
curves comparing the number of sequences with the number of OTUs found in the 16S rRNA gene libraries from the microbiota in the feces of the
mice in the five groups; (b) alpha diversity (Pielou’s e) illustrating the diversity of each group; (c) Venn diagrams of OTUs demonstrating overlap
among the groups; (d and e) beta diversity (PCA and NMDS) illustrating the diversity of each group; (f ) the top 16 phylum-level stacked-bar; (g) the
top 30 genus-level stacked-bar; (h) histogram of LDA scores >2 for p_phylum, c_class, o_order, f_family, g_genus and s_species; and (i) taxonomic
cladogram. Branching the evolutionary tree diagram, the circles radiating from inside to outside in the diagram represent the taxonomic levels from
phylum to genus (the innermost one with yellow circles is the boundary). Each small circle at a different taxonomic level represents a taxon at that
level, and the diameter size of the small circles represents the relative abundance size. ( j) The relative abundances of Firmicutes, Bacteroidota, the
ratio of Firmicutes to Bacteroidota, Verrucomicrobiota, Bacteroides, Escherichia–Shigella and Desulfovibrionaceae in the Ctrl, DSS, DSS + LA and
DSS + V. ratti, DSS + mixture groups. Compared with the Ctrl group, **p < 0.01; compared with the DSS group, ##p < 0.01; DSS + V. ratti vs. DSS +
mixture, *p < 0.05.

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function


Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Fig. 9 V. ratti and LA regulated the production of lactate, SCFAs, and SCFA-producing bacteria. (a) The levels of acetic acid, propionic acid, isobuty-
ric acid, butyric acid, isovaleric acid and valeric acid; (b) the levels of total SCFAS; (c) the production of lactate in the feces; (d) the relative abundance
of acetate-producing bacteria; and (e) the relative abundance of butyrate-producing bacteria. Data are presented as mean ± SD, n = 5/group.
Significant differences were calculated by one-way ANOVA, followed by Tukey’s test for multiple comparisons,*p < 0.05; **p < 0.01; ***p < 0.001;
****p < 0.0001, compared to the Ctrl group; #p < 0.05; ##p < 0.01; ##p < 0.001; ####p < 0.0001, compared to the DSS group.

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper

combination of V. ratti and LA in a DSS-induced colitis and converting it primarily into acetate and propionate.40 In
mouse model. our previous study, we conducted experiments adjusting the
As expected, the co-administration of V. ratti and LA posi- glucose and sodium lactate concentrations to 4.0 g L−1 and
tively influenced the intestinal microenvironment, leading to 4.5 g L−1, respectively, based on medium 136, to assess the
an improved colitis status in mice. The intervention with yield of SCFAs in the medium after 24 hours of co-culture. The
V. ratti and LA yielded multiple beneficial outcomes, including results demonstrated a significant increase, approximately
an increase in colon length, a reduction in disease activity, tenfold, in both acetic and propionic acid production during
lower disease activity index (DAI) scores, elevated levels of anti- co-culture (data unpublished). To further explore the dynamics
oxidant factors, reduced expression of pro-inflammatory of cross-feeding between V. ratti and LA under conditions with
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

factors, and relief from colitis symptoms. Notably, the combi- limited carbon sources, we reduced the amount of trypsin and
nation of V. ratti and LA demonstrated enhanced efficacy com- yeast dip powder by five-fold, removed sodium lactate, and
pared to administering mice with them separately. These find- increased glucose to 8.0 g L−1. Our experimental data showed
ings suggest that this novel probiotic blend holds promise as a that V. ratti and LA in co-culture primarily produced acetic
potential treatment for alleviating inflammatory bowel disease. acid, propionic acid, and butyric acid, all of which were at sig-
Oxidative stress and immune response are key factors in nificantly higher levels compared to mono-cultures. These
tissue damage associated with UC.29,30 The excessive pro- findings suggest the mutual promotion of proliferation by
duction of reactive oxygen species (ROS) disrupts the balance V. ratti and LA through cross-feeding during co-culture,
between oxidants and antioxidants, resulting in oxidative wherein LA rapidly converts glucose into lactate, which, as an
damage to the host.31 In our experimental study, we observed available carbon source for V. ratti, is further utilized to
a significant increase in myeloperoxidase (MPO) accumulation produce SCFAs.
in the colon tissue of mice in the DSS group, indicating heigh- Short-chain fatty acids (SCFAs), including acetate, propio-
tened levels of inflammation. However, the co-administration nate, and butyrate, play a crucial role in maintaining intestinal
of V. ratti and LA significantly reduced MDA and MPO pro- homeostasis and serve as an important fuel for intestinal epi-
duction, improved SOD activity, and decreased GSH levels in thelial cells, strengthening the integrity of the gut barrier.2,14
DSS-challenged mice (Fig. 6). These findings underscore the These SCFAs have exhibited benefits in preventing intestinal
potential of the V. ratti and LA combination to mitigate the diseases, including IBD.41–43 Moreover, previous research has
inflammatory responses in the colon triggered by DSS, enhan- highlighted their capacity to inhibit the proliferation of patho-
cing antioxidant activity and decreasing oxidative damage. gens within the host’s intestinal microenvironment.44
UC is often characterized by uncontrolled and highly acti- In order to gain deeper insights into how this probiotic
vated inflammation within the intestinal mucosa.32 Previous combination influences the intestinal microflora and their
studies have reported elevated iNOS expression in the colonic metabolites, we conducted analyses of the gut microbiota and
mucosal epithelium and crypts of individuals suffering from metabolites (including SCFAs and lactic acid) in the UC mice
ulcerative colitis (UC), which has been confirmed through subjected to treatment with V. ratti and LA.
in vitro cellular experiments.33,34 Moreover, pro-inflammatory In our study, we observed a significant enrichment of
cytokines such as IL-1β, IL-γ, IL-6, IL-17a, and IFN-γ are com- Ligilactobacillus in the mixture group during the intervention
monly involved in initiating intestinal inflammation in UC.35 period. Ligilactobacillus salivarius is known for its anti-
In our study, DSS treatment increased the expression of these microbial and immune-regulatory properties, which contribute
pro-inflammatory cytokines, while the co-administration of to the regulation of intestinal microbiota.45,46 We speculate
V. ratti and LA significantly regulated their levels (Fig. 5). We that the one-week intervention with V. ratti and LA in mice
speculate that this combination may alleviate colitis by inhibit- effectively increased the relative abundance of the beneficial
ing the signaling pathway and activating the NRF-2 signaling bacterium Ligilactobacillus, thereby preventing the develop-
pathway, both of which play roles in regulating inflammatory ment of colitis. Furthermore, during the UC model induction
responses.36,37 Disruption of the intestinal epithelial barrier period, the DSS group displayed a significant increase in the
function is also implicated in UC.38 We analyzed the relative abundance of pro-inflammatory bacteria such as
expression of proteins that maintain the intestinal barrier Desulfovibrio, Escherichia–Shigella, and Helicobacter, contribut-
function and found that V. ratti combined with LA positively ing to greater intestinal inflammation. In contrast, the DSS +
modulated the DSS-induced decrease in the expression of mixture group exhibited an increase in the relative abundance
barrier-maintaining proteins such as MUC2, MUC3 and occlu- of beneficial bacteria, including Akkermansia, Ruminococcus,
din in mice. The results indicate that V. ratti combined with and Eubacterium, which are known for their capacity to
LA is able to restore epithelial barrier integrity and relieve UC produce SCFAs (Fig. 9D and E). And their increased abundance
symptoms. in the DSS + mixture group corresponded to higher levels of
Bacteria engage in both competition for nutrients and SCFAs.47,48 The above findings suggest that the intervention
cooperative processes such as metabolic cross-feeding, where with V. ratti and LA reversed the microbiota alterations and
one strain’s metabolites serve as nutrition for another.39 alleviated the inflammatory responses in mice.
Previous studies conducted in the rumen and the pig GI tract The microbial community of the human colon contains
have established that Veillonella are capable of utilizing lactate numerous lactic acid-producing bacteria. However, lactate is

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function

normally detected at low concentrations (<5 mM) in feces from Ethical approval
healthy individuals.22,49 In our study, the DSS group exhibited
a significantly higher lactic acid content than the Ctrl group. The animals were allowed free access to standard chow and
Interestingly, the DSS + mixture group exhibited a lower lactic sterilized water. All experiments involving animals were
acid content compared to both the DSS and DSS + V. ratti approved by the Animal Ethics and Welfare Committee of the
groups. This finding indicates that LA promotes the growth of Northwest A&F University, China (SP2022015).
V. ratti, facilitating efficient consumption of lactic acid and its
conversion into SCFAs. As anticipated, the DSS + mixture
group demonstrated higher levels of total SCFAs in the feces. Data availability
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Collectively, the colonization with V. ratti and LA in mice


resulted in increased SCFA levels. This modulation of the gut The datasets analyzed during the current study are available
microbiota and metabolites contributed to the maintenance of from the corresponding author on reasonable request.
intestinal barrier integrity and control of intestinal inflam-
mation through enhanced immune functions. Therefore, the
administration of combined V. ratti and LA promoted the pro- Conflicts of interest
duction of SCFAs, which played a vital role in regulating intes-
There are no conflicts to declare.
tinal health.
This study has several limitations that should be acknowl-
edged. First, we are yet to delve deeply into the workings of the
mechanisms of inflammatory and antioxidant factors that
Acknowledgements
underlie the regulatory effects of V. ratti and LA in chronic UC. This work was supported by Grant No. 31972043 from the
Second, while our results are promising, the application of National Natural Science Foundation of China and Grant
V. ratti and LA as a probiotic combination in UC patients 2023-YBNY-146 from the General project – agricultural field.
requires rigorous safety evaluation and clinical intervention
trials to confirm the results observed in our animal model.
Third, our focus on lactic acid and short-chain fatty acids rep- References
resents only a small fraction of the potential intestinal metab-
olites influenced by V. ratti and LA. We recognize the need for 1 A. Jentzer, P. Veyrard, X. Roblin, P. Saint-Sardos,
more comprehensive investigations on the broader spectrum N. Rochereau, S. Paul, T. Bourlet, B. Pozzetto and S. Pillet,
of intestinal metabolites in future studies. Cytomegalovirus and Inflammatory Bowel Diseases (IBD)
with a Special Focus on the Link with Ulcerative Colitis
(UC), Microorganisms, 2020, 8, 1078.
2 D. Parada Venegas, M. K. De la Fuente, G. Landskron,
5. Conclusion M. J. González, R. Quera, G. Dijkstra, H. J. M. Harmsen,
K. N. Faber and M. A. Hermoso, Short Chain Fatty Acids
The co-administration of V. ratti and LA positively impacted
(SCFAs)-Mediated Gut Epithelial and Immune Regulation
the intestinal microenvironment, resulting in notable improve-
and Its Relevance for Inflammatory Bowel Diseases, Front.
ments in the colitis status of mice. This was evident through
Immunol., 2019, 10, 3389.
several key observations: an increase in colon length, a
3 S. Kanwal, T. P. Joseph, S. Aliya, S. Song, M. Z. Saleem,
reduction in disease activity, and enhanced antioxidant
M. A. Nisar, Y. Wang, A. Meyiah, Y. Ma and Y. Xin,
activity. These findings underscore promising therapeutic
Attenuation of DSS induced colitis by Dictyophora indu-
potential of this novel probiotic combination for mitigating
siata polysaccharide (DIP) via modulation of gut microbiota
inflammatory bowel disease.
and inflammatory related signaling pathways, J. Funct.
Foods, 2020, 64, 103641.
4 F. Genua, V. Raghunathan, M. Jenab, W. M. Gallagher and
Author contributions D. J. Hughes, The Role of Gut Barrier Dysfunction and
Microbiome Dysbiosis in Colorectal Cancer Development,
Na Li: conceptualization, methodology, and writing – original Front. Oncol., 2021, 11, 626349.
draft. Hejing Wang: investigation, data curation and software. 5 J.-C. Xue, S. Yuan, H. Meng, X.-T. Hou, J. Li, H.-M. Zhang,
Huizhu Zhao: methodology, software and validation. L.-L. Chen, C.-H. Zhang and Q.-G. Zhang, The role and
Mengyang Wang: visualization, investigation and formal ana- mechanism of flavonoid herbal natural products in ulcera-
lysis. Yi Hao: data curation. Jia Yu: conceptualization and tive colitis, Biomed. Pharmacother., 2023, 158, 114086.
resources. Jin Cai: methodology and software. Yun Jiang: 6 C. Milani, S. Duranti, F. Bottacini, E. Casey, F. Turroni,
writing – review and editing. Xin Lü: supervision. Bianfang J. Mahony, C. Belzer, S. D. Palacio, S. A. Montes,
Liu: supervision and funding acquisition. All authors have L. Mancabelli, G. A. Lugli, J. M. Rodriguez, L. Bode,
read and agreed to the published version of the manuscript. W. D. Vos, M. Gueimonde, A. Margolles, D. V. Sinderen and

Food Funct. This journal is © The Royal Society of Chemistry 2023


View Article Online

Food & Function Paper

M. Ventura, The First Microbial Colonizers of the Human 19 E. A. Delwiche, J. J. Pestka and M. L. Tortorello, The veillo-
Gut: Composition, Activities, and Health Implications of nellae: Gram-negative cocci with a unique physiology,
the Infant Gut Microbiota, Microbiol. Mol. Biol. Rev., 2017, Annu. Rev. Microbiol., 1985, 39, 175–193.
81, e00036-17. 20 Z. Huang, X. Zhou, C. Stanton, R. P. Ross, J. Zhao,
7 J. Lloyd-Price, G. Abu-Ali and C. Huttenhower, The healthy H. Zhang, B. Yang and W. Chen, Comparative Genomics
human microbiome, Genome Med., 2016, 8, 51. and Specific Functional Characteristics Analysis of
8 W. Lv, D. Zhang, T. He, Y. Liu, L. Shao, Z. Lv, X. Pu, Y. Wang Lactobacillus acidophilus, Microorganisms, 2021, 9, 9.
and L. Liu, Combination of Lactobacillus plantarum improves 21 S. H. Duncan, P. Louis and H. J. Flint, Lactate-Utilizing
the effects of tacrolimus on colitis in a mouse model, Front. Bacteria, Isolated from Human Feces, That Produce
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

Cell. Infect. Microbiol., 2023, 13, 1130820. Butyrate as a Major Fermentation Product, Appl. Environ.
9 Q.-Y. Cui, X.-Y. Tian, X. Liang, Z. Zhang, R. Wang, Y. Zhou, Microbiol., 2004, 70, 5810–5817.
H.-X. Yi, P.-M. Gong, K. Lin, T.-J. Liu and L.-W. Zhang, 22 L. Chen, R. Li, Z. Wang, Z. Zhang, J. Wang, Y. Qiao,
Bifidobacterium bifidum relieved DSS-induced colitis in Y. Huang and W. Liu, Lactate-utilizing bacteria ameliorates
mice potentially by activating the aryl hydrocarbon recep- DSS-induced colitis in mice, Life Sci., 2022, 288, 120179.
tor, Food Funct., 2022, 13, 5115–5123. 23 B. Liu, X. Yin, H. Yu, Y. Feng, X. Ying, J. Gong and
10 W.-K. Kim, D. H. Han, Y. J. Jang, S. Park, S. J. Jang, G. Lee, C. L. Gyles, Alteration of the Microbiota and Virulence
H. S. Han and G. Ko, Alleviation of DSS-induced colitis via Gene Expression in E. coli O157:H7 in Pig Ligated Intestine
Lactobacillus acidophilus treatment in mice, Food Funct., with and without AE Lesions, PLoS One, 2015, 10,
2021, 12, 340–350. e0130272.
11 X. Li, Y. Yang and B. Zhang, Lactate metabolism in human 24 R. Mennigen, K. Nolte, E. Rijcken, M. Utech, B. Loeffler,
health and disease, Signal Transduction Targeted Ther., N. Senninger and M. Bruewer, Probiotic mixture VSL#3 pro-
2022, 7, 305. tects the epithelial barrier by maintaining tight junction
12 M. Kraatz, R. J. Wallace and L. Svensson, Olsenella umbo- protein expression and preventing apoptosis in a murine
nata sp. nov., a microaerotolerant anaerobic lactic acid bac- model of colitis, Am. J. Physiol.: Gastrointest. Liver Physiol.,
terium from the sheep rumen and pig jejunum, and 2009, 296, G1140–G1149.
emended descriptions of Olsenella, Olsenella uli and 25 M. Murano, K. Maemura, I. Hirata, K. Toshina,
Olsenella profusa, Int. J. Syst. Evol. Microbiol., 2011, 61, T. Nishikawa, N. Hamamoto, S. Sasaki, O. Saitoh and
795–803. K. Katsu, Therapeutic effect of intracolonically adminis-
13 D. I. Poppleton, M. Duchateau, V. Hourdel, M. Matondo, tered nuclear factor κB ( p65) antisense oligonucleotide on
J. Flechsler, A. Klingl, C. Beloin and S. Gribaldo, Outer mouse dextran sulphate sodium (DSS)-induced colitis, Clin.
Membrane Proteome of Veillonella parvula: A Diderm Exp. Immunol., 2001, 120, 51–58.
Firmicute of the Human Microbiome, Front. Microbiol., 26 H. Melhem, M. R. Spalinger, J. Cosin-Roger, K. Atrott,
2017, 8, 01215. S. Lang, K. A. Wojtal, S. R. Vavricka, G. Rogler and I. Frey-
14 S. Deleu, K. Machiels, J. Raes, K. Verbeke and S. Vermeire, Wagner, Prdx6 Deficiency Ameliorates DSS Colitis:
Short chain fatty acids and its producing organisms: An Relevance of Compensatory Antioxidant Mechanisms,
overlooked therapy for IBD?, EBioMedicine, 2021, 66, J. Crohns Colitis, 2017, 11, 871–884.
103293. 27 S. Zhu, M. Han, S. Liu, L. Fan, H. Shi and P. Li,
15 S. Fu, Y. Yuan, X. Tian, L. Zhou, L. Guo, D. Zhang, J. He, Composition and diverse differences of intestinal micro-
C. Peng, Y. Qiu, C. Ye, Y. Liu and B. Zong, Detection of biota in ulcerative colitis patients, Front. Cell. Infect.
Colistin Sulfate on Piglet Gastrointestinal Tract Microbiol., 2022, 12, 953962.
Microbiome Alterations, Vet. Sci., 2022, 9, 666. 28 C. McShane, R. Corcoran, F. O’Connell, P. McDonagh,
16 N. M. Quijada, R. Bodas, J. M. Lorenzo, S. Schmitz-Esser, J. O’Sullivan and D. Kevans, P089 Colonic explant lactate
D. Rodríguez-Lázaro and M. Hernández, Dietary concentration and inflammatory protein secretion in
Supplementation with Sugar Beet Fructooligosaccharides ulcerative colitis, J. Crohns Colitis, 2023, 17, i251–i253.
and Garlic Residues Promotes Growth of Beneficial 29 S. Quattrini, S. Gatti, A. Palpacelli, S. Mattioli,
Bacteria and Increases Weight Gain in Neonatal Lambs, L. Cianfruglia, T. Galezzi, C. Monachesi, C. Quatraccioni,
Biomolecules, 2020, 10, 1179. G. Catassi, A. Di Sario, T. Armeni and C. Catassi, P549
17 J. A. Durant, D. J. Nisbet and S. C. Ricke, Response of Oxidative stress and antioxidant capacity biomarkers in
selected poultry cecal probiotic bacteria and a primary adults and children with IBD: current update from
poultry salmonella typhimurium isolate grown with or OxIBDiet trial, J. Crohns Colitis, 2022, 16, i496–i496.
without glucose in liquid batch culture, J. Environ. Sci. 30 E. Dudzińska, M. Gryzinska, K. Ognik, P. Gil-Kulik and
Health, Part B, 2000, 35, 503–516. J. Kocki, Oxidative Stress and Effect of Treatment on the
18 M. Kraatz and D. Taras, Veillonella magna sp. nov., isolated Oxidation Product Decomposition Processes in IBD, Oxid.
from the jejunal mucosa of a healthy pig, and emended Med. Cell. Longevity, 2018, 2018, 7918261.
description of Veillonella ratti, Int. J. Syst. Evol. Microbiol., 31 Y. Dincer, Y. Erzin, S. Himmetoglu, K. N. Gunes, K. Bal and
2008, 58, 2755–2761. T. Akcay, Oxidative DNA Damage and Antioxidant Activity

This journal is © The Royal Society of Chemistry 2023 Food Funct.


View Article Online

Paper Food & Function

in Patients with Inflammatory Bowel Disease, Dig. Dis. Sci., 41 Z. Zhang, H. Zhang, T. Chen, L. Shi, D. Wang and D. Tang,
2007, 52, 1636–1641. Regulatory role of short-chain fatty acids in inflammatory
32 J. H. Park, L. Peyrin-Biroulet, M. Eisenhut and J. I. Shin, IBD bowel disease, Cell Commun. Signaling, 2022, 20, 64.
immunopathogenesis: A comprehensive review of inflamma- 42 M. Akhtar, Y. Chen, Z. Ma, X. Zhang, D. Shi, J. A. Khan and
tory molecules, Autoimmun. Rev., 2017, 16, 416–426. H. Liu, Gut microbiota-derived short chain fatty acids are
33 M. Cavicchi, L. Gibbs and B. J. Whittle, Inhibition of indu- potential mediators in gut inflammation, Anim. Nutr.,
cible nitric oxide synthase in the human intestinal epi- 2022, 8, 350–360.
thelial cell line, DLD-1, by the inducers of heme oxygenase 43 C. Li, Y. Zhao, J. Cheng, J. Guo, Q. Zhang, X. Zhang, J. Ren,
1, bismuth salts, heme, and nitric oxide donors, Gut, 2000, F. Wang, J. Huang, H. Hu, R. Wang and J. Zhang, A
Published on 19 October 2023. Downloaded by University of Toronto on 11/27/2023 5:05:11 AM.

47, 771–778. Proresolving Peptide Nanotherapy for Site-Specific


34 H. Kleinert, A. Pautz, K. Linker and P. M. Schwarz, Treatment of Inflammatory Bowel Disease by Regulating
Regulation of the expression of inducible nitric oxide Proinflammatory Microenvironment and Gut Microbiota,
synthase, Eur. J. Pharmacol., 2004, 500, 255–266. Adv. Sci., 2019, 6, 1900610.
35 D. Chauhan, L. Van de Walle and M. Lamkanfi, 44 Y. Tian, Q. Xu, L. Sun, Y. Ye and G. Ji, Short-chain fatty acids
Therapeutic modulation of inflammasome pathways, administration is protective in colitis-associated colorectal
Immunol. Rev., 2020, 297, 123–138. cancer development, J. Nutr. Biochem., 2018, 57, 103–109.
36 K. Ha Kim, R. T. Sadikot, J. Yeon Lee, H.-S. Jeong, Y.-K. Oh, 45 B. D. Chaves, M. M. Brashears and K. K. Nightingale,
T. S. Blackwell and M. Joo, Suppressed ubiquitination of Applications and safety considerations of Lactobacillus sali-
Nrf2 by p47phox contributes to Nrf2 activation, Free varius as a probiotic in animal and human health, J. Appl.
Radicals Biol. Med., 2017, 113, 48–58. Microbiol., 2017, 123, 18–28.
37 T. Liu, L. Zhang, D. Joo and S.-C. Sun, NF-κB signaling in 46 L. Shang, H. Liu, H. Yu, M. Chen, T. Yang, X. Zeng and
inflammation, Signal Transduction Targeted Ther., 2017, 2, S. Qiao, Core Altered Microorganisms in Colitis Mouse
17023. Model: A Comprehensive Time-Point and Fecal Microbiota
38 J. Mankertz and J.-D. Schulzke, Altered permeability in Transplantation Analysis, Journal, 2021, 10, 643.
inflammatory bowel disease: pathophysiology and clinical 47 H. Zeng, C. Huang, S. Lin, M. Zheng, C. Chen, B. Zheng
implications, Curr. Opin. Gastroenterol., 2007, 23, 379–383. and Y. Zhang, Lotus Seed Resistant Starch Regulates Gut
39 A. Belenguer, S. H. Duncan, A. G. Calder, G. Holtrop, Microbiota and Increases Short-Chain Fatty Acids
P. Louis, G. E. Lobley and H. J. Flint, Two Routes of Production and Mineral Absorption in Mice, J. Agric. Food
Metabolic Cross-Feeding between Bifidobacterium adoles- Chem., 2017, 65, 9217–9225.
centis and Butyrate-Producing Anaerobes from the Human 48 L. E. M. Willemsen, M. A. Koetsier, S. J. H. van Deventer,
Gut, Appl. Environ. Microbiol., 2006, 72, 3593–3599. et al., Short chain fatty acids stimulate epithelial mucin 2
40 T. Sato, K. Matsumoto, T. Okumura, W. Yokoi, E. Naito, expression through differential effects on prostaglandin
Y. Yoshida, K. Nomoto, M. Ito and H. Sawada, Isolation of E(1) and E(2) production by intestinal myofibroblasts, Gut,
lactate-utilizing butyrate-producing bacteria from human 2003, 52, 1442–1447.
feces and in vivo administration of Anaerostipes caccae 49 H. Hove, M. R. Clausen and P. B. Mortensen, Lactate and
strain L2 and galacto-oligosaccharides in a rat model, pH in faeces from patients with colonic adenomas or
FEMS Microbiol. Ecol., 2008, 66, 528–536. cancer, Gut, 1993, 34, 625.

Food Funct. This journal is © The Royal Society of Chemistry 2023

You might also like