You are on page 1of 11

Biomedicine & Pharmacotherapy 125 (2020) 109914

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


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

Composite probiotics alleviate type 2 diabetes by regulating intestinal T


microbiota and inducing GLP-1 secretion in db/db mice
Yanming Wang1, Dinareer Dilidaxi1, Yuche Wu, Jialehasibieke Sailike, Xin Sun, Xin-hua Nabi*
Department of Pharmacology, Xinjiang Medical University, Urumqi, 830011, China

ARTICLE INFO ABSTRACT

Keywords: Backgroud/Aim: Previous studies have found that probiotic fermented camel milk has anti-diabetic effect by
Diabetes 2 mellitus inducing (glucagon-like peptide-1) GLP-1 secretion. Probiotics are valuable in prevention and treatment of
Composite probiotics diabetes. As a result, our team islolated 14 probiotics from fermented camel milk. These probiotics have ben-
Gut microbiota eficial characteristics, but the possible anti-diabetic mechanisms remains unclear. The present study aimed to
Short-chain fatty acids
explore the possoble anti-diabetic mechanisms of 14 probiotics.
Glucagon-like peptide-1
Methods: C57BL/Ks mice were normal group. The db/db mice were randomized into five groups: model group,
metformin group, liraglutide group, low-dose and high-dose probiotic group. Biochemical parameters were
determined by the respective assay kits. The levels of the short-chain fatty acids (SCFAs) and microbiota were
respectively determined by gas chromatography and qRT-PCR. HE staining and immunofluorescence were used
for histomorphological observation. Quantitative PCR and western-blot were determined the gene and protein
expression of Bax, Bcl-2, Caspase-3 and PI3K/AKT.
Results: Probiotics significantly improved blood glucose and blood lipid parameters, as well as the morpholo-
gical changes of pancreas, liver and kidney. Probiotics improved the gut barrier function through increasing the
levels of SCFA-producing bacteria and SCFAs as well as the expression of claudin-1 and mucin-2, and decreasing
Escherichia coli and LPS level. In additon, probiotics enhanced insulin secretion through glucose-triggered GLP-1
secretion by upregulating G protein-coupled receptor 43/41 (GPR43/41), proglucagon and proconvertase 1/3
activity. Forthermore, probiotics protected pancreas against apoptosis, which may be dependent on the upre-
gulation of PI3K/AKT pathway.
Conclusions: The anti-diabetic effect of 14 probiotics in db/db mice seem to be related to an increase of SCFA-
producing bacteria, the improvement of intestinal barrier function and the upregulation of GLP-1 production,
and indicate these probiotics might be a good candidate to prevent and treat diabetes.

1. Introduction is benefical to control blood glucose levels due to the downregulation


level of insulin in later stages of T2D [3]. Glucagon-like peptide-1 (GLP-
The prevalence of diabetes mellitus (DM), a metabolic disorder that 1) is a hormone and secreted by intestinal L cells loacted mainly in the
affects global health, shows a rising trend in both developed and de- distal ileum and colon. The absorption of nutrients, especially the
veloping countries [1]. The data of the World Health Organization glucose or carbohydrates, can stimulate GLP-1 secretion [4]. GLP-1 can
(WHO) shows that the incidence of diabetes continues to rise year by normalize the glucose homeostasis by enhancing and sensitizing β cells
year. The world prevalence of adult diabetes in 2010 is 285 million and function in response to glucose-stimulated insulin secretion [5]. GLP-1
predicted to reach 439 million in 2030 [2]. Notably, T2D comprises 80 also plays an important role in controlling glucose homeostasis by sti-
% of diabetes and is characterised by a combination of inadequate in- mulating pancreatic β-cells to release insulin, thereby reducing the se-
sulin secretion and insulin resistance along with hyperglycaemia, dys- cretion of glucagon in pancreatic α-cells and inhibiting the appetite [6].
lipidemia and glucose intolerance. Especially additional insulin therayp Besides, GLP-1 also inhibits pancreatic β-cell apoptosis and contributes

Abbreviations: GLP-1, Glucagon-like peptide-1; GCG, Proglucagon; PC1/3, Proconvertase 1/3; TNF-α, Tumor necrosis factor α; IFN-γ, Interferon-γ; IL-1β,
Interleukin-1β; LPS, Lipopolysaccharide; GPR43/41, G protein-coupled receptor 43/41; PI3K/AKT, Phosphatidylinositol 3ʹ-kinase/protein kinase B; Bcl-2, B-cell
lymphoma 2; Bax, Bcl-2-associated X protein; SCFAs, Short-chain fatty acids

Corresponding author.
E-mail address: xinhuanabi@yeah.net (X.-h. Nabi).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biopha.2020.109914
Received 5 October 2019; Received in revised form 10 January 2020; Accepted 12 January 2020
0753-3322/ © 2020 The Author(s). Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

to β-cell growth [7]. Therefore, GLP-1 might be as one of the potential 05). The experimental protocol complied with the ARRIVE guidelines
therapies in T2D by improving the pancreatic beta-cells function and and was carried in compliance with National Institutes of Health guide
inducing insulin secretion. for the care and use of Laboratory animals (NIH Publications No. 8023,
There is increasing evidence showing that the relationship between revised 1978). The specific pathogen-free (SPF) grade male db/db mice
gut microbiota and diabetes is intertwined. Gut microorganisms are (∼35 g average bodyweight) and C57BL/Ks mice (∼20 g average
part of an extremely sophisticated ecosystem. There are abundant, bodyweight) were at 6 weeks of age [19]. Normal chow diet and water
multifarious microorganisms living in the intestines, such as were provided ad libitum. The mice were housed in groups of three to
Fusobacteria, Actinobacteria, Firmicutes, and Bacteroidetes. Recent studies four per cage at 22 ℃ ± 1 ℃ with 50 % ± 5 % humidity and a 12-h
have shown that the composition and abundance of gut microbiota in diurnal light cycle. The mice were acclimated for 1 week before the
healthy individuals are remarkably different from patients with T2D experiment. C57BL/Ks mice were the normal (N) control group (n =
[8]. The short-chain fatty acid (SCFA)-producing bacteria can produce 10) and orally fed physiological saline (0.5 mL/d) for 6 weeks. Diabetic
SCFAs (e.g., acetic acid, propionic acid, butyric acid) by the digestion, db/db mice were randomly divided into five groups: the diabetic model
fermentation and catabolism of nondigestible dietary fibers, proteins, (D) group (n = 10) was orally fed physiological saline (0.5 mL/d) for 6
and glycoproteins. Studies in humans have reported that levels of bu- weeks; the positive control groups (n = 10/group) received respec-
tyrate-producing bacteria in T2D patients are significantly decreased tively intragastric administration of metformin (M) (0.3 g/kg/d) (Novo
compared to healthy individuals [9]. Animal study has described si- Nordisk Biotechnology Co Ltd., Denmark, cat# JVGR828-1) and in-
milar results indicating that the levels of SCFA-producing bacteria in jection of liraglutide (L) (0.2 μg/g/d) (Sino-American Shanghai Squibb
T2D model are significantly decreased compared to normal group [10]. Pharmaceutical Co., Ltd., Shanghai, China, cat# ABK5947) for 6 weeks
The diabetic prevalence of Kazakh populations was lower than other [20,21]. The probiotic groups (n = 10/group) were orally fed with the
ethnic groups in Xinjiang of China, which has a colse relationship with low-dose of probiotics and high-dose probiotics once a day for 6 weeks
their custom of daily use of fermented dairy products for a long time, [22,23]. The low-dose of compounded probiotics in the 0.5 mL phy-
such as fermented camel milk [11–13]. In the previous study, we have siological saline includes 10 Lactobacillus strains (1 × 108 CFU/mL) and
found that the probiotic fermented camel milk has anti-diabetic effect 4 saccharomycetes (1 × 106 CFU/mL). The high-dose of compounded
by inducing GLP-1 secretion in streptozotocin-induced DM rats [14]. probiotics in the 0.5 mL physiological saline includes 10 Lactobacillus
Emerging evidence has shown that probiotics are expected to be a strains (1 × 1010 CFU/mL) and 4 saccharomycetes (1 × 108 CFU/mL).
promising therapeutic and preventive strategy for diabetes mellitus by All mice were given intervention at 11 o’clock in the morning once a
modifying intestinal microbiota, improving intestinal barrier function day for 6 weeks.
and inhibiting insulin resistance [15]. Therefore, 14 probiotics were
isolated from Xinjiang traditional fermented camel milk and identified 2.3. Fasting blood glucose (FBG) and Oral glucose tolerance test (OGTT)
by the Center for the Preservation of Chinese Industrial Microbial
Strains [14]. These probiotics have beneficial characteristics, including The body weights of mice in different groups were recorded on the
acid resistance, bile tolerance, high self-aggregation ability, high ad- first day of treatment administration and then thrice weekly until 6
hesion to Caco-2 cells and so on [16,17]. However, the possible anti- weeks. For the fasting blood-glucose, the mice were fasted for 8 h from
diabetic mechanisms associated with these probiotics remain unclear. 10 o’clock in the morning to 6 o'clock in the afternoon at the Sunday of
The study aimed to explore the possible mechanisms involved in pro- every week for 6 weeks. The tail was sterilized by the alcohol sponge
biotic-induced GLP-1 secretion in db/db mice. and made a small cut at the tip of tail vein. The first drop of blood was
thrown away and then the second drop of blood was detected by the
2. Materials and methods glucometer (Roche Diagnostics, Mannheim, Germany). After being in-
tervention for 6 weeks, all mice were starved for 8 h and oral glucose
2.1. Probiotic growth and preparation tolerance tests were carried out using 2 g/kg body weight of glucose (20
% solution). We collected the tail blood samples at 0, 30, 60, 90 and
Fourteen probiotics were isolated from traditional fermented camel 120 min after glucose loading and detected the blood glucose value by
milk in Xinjiang, China and identified by the Center for the Preservation the glucometer (Roche Diagnostics, Mannheim, Germany). The area
of Chinese Industrial Microbial Strains [14]. Probiotics were cultured under the glucose curve (AUC glucose) was determined from time
according to a previously reported method [18]. Briefly, ten Lactoba- 0–120 min (AUC 0–120 min) after glucose administration.
cillus strains were cultured in Man Rogosa Sharpe (MRS) broth at 37 ℃
for 48 h. Four yeast strains were cultured in modified-MRS broth at 37 2.4. ELISA determination of total GLP-1 and insulin
℃ for 48 h. Cultured cells were harvested by centrifugation at 3000 rpm
(2570 × g) for 15 min and then washed twice with aseptic phosphate Mice were performed by OGTT after fasting for 8 h. At 0 min, 15
buffered saline. The high-dose of probiotics includes 1 × 1010 colony- min, 30 min and 45 min after glucose load, the blood collected from the
forming units (CFU)/mL of ten Lactobacillus strains (Lactobacillus plan- tail veins was immediately transferred into EDTA tubes containing a
tarum, Lactobacillus helveticus, Lactococcus lactis, Lactobacillus pentosus, dipeptidyl peptidase IV inhibitor (EMD Millipore, United States), and
Lactobacillus paracasei, Lactobacillus paracasei sbusp.tolerans, Lactoba- then centrifuged at 1200 g for 15 min at 4 ℃. The plasma was im-
cillus mucosae, Lactobacillus rhamnosus, Lactobacillus harbinensis and mediately dispensed into a new tube and subjected to enzyme im-
Lactobacillus hilgardii) and 1 × 108 CFU/mL of 4 yeast strains (Is- munoassay. The total GLP-1 and insulin levels in plasma were detected
satchenkia orientalis, Candida ethanolica, Kluyveromyces marxianus and by GLP-1 (Mesoscale, Rockville, MD) and insulin (Uscn life, Wuhan,
Pichia membranifaciens). The low-dose of probiotics includes 1 × 108 China) ELISA assays according to kit instructions.
colony-forming units (CFU)/mL of ten Lactobacillus strains and 1 × 106
CFU/mL of 4 yeast strains. 2.5. Biochemical parameter analysis

2.2. Animals and treatments The triglyceride (TG), total cholesterol (TC), high density lipopro-
tein cholesterol (HDL-C), low density lipoprotein cholesterol (LDL-C)
Specific pathogen free (SPF) grade male db/db mice and C57BL/KS and lipopolysaccharide (LPS) levels were determined by enzyme-linked
male mice were purchased from the Experimental Animal Center at immunosorbent assay (ELISA) kits (Nanjing Jiancheng Biology Co.,
Xinjiang Medical University. The study was approved by the institu- Ltd., Nanjing, China) according to the kit instructions. Haemoglobin
tional animal ethics committee (Approval Number: IACUC20160414- A1c (HbA1c) and C-peptide were detected by the ELISA kits (Wuhan

2
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

Table 1 2.8. Haematoxylin and eosin (H&E) examination and


Sequences of primers used in bacterial expression analysis. immunohistochemistry
Target Forward and reverse primers Reference
Pancreas, liver, kidney and colon tissues were collected and fixed in
Lactobacillus F: AGCAGTAGGGAATCTTCCA [36] 10 % formalin saline for 72 h, dewatered in different concentrations of
R: CACCGCTACACATGGAG
alcohol and embedded in paraffin. Samples were sectioned at 5 μm and
Bifidobacterium F: GTCAGCTCGTGTCGTGAG [36]
R: GTCGCATCCCGTTGTACC
stained with haematoxylin and eosin. Pancreas and colon tissues were
Clostridium leptum F: GCACAAGCAGTGGAGT [36] placed into an incubator for 2 h at 70 ℃, followed by administration of
R: CTTCCTCCGTTTTGTCA dimethylbenzene, different concentrations of ethylalcohol, distilled
Roseburia F: GCGGTRCGGCAAGTCTGA [28] water, 3 % hydorgen peroxide and citric acid. Pancreas or colon tissues
R: CCTCCGACACTCTAGTMCGAC
were incubated with 5 % BSA at 37 ℃ for 30 min and incubated with
Fusobacterium F: GTATGTCRCAAGCGTTATCC [28]
R: AACGCAATACRGAGTTGAGC anti-claudin-1 (Abcam, cat# ab15098, 1:50), anti-insulin (Abcam, cat#
Prevotella F: CGAACAGGATTAGATACCC [28] ab181547, 1:1000), anti-Bax (Abcam, cat# ab32503, 1:100), anti-Bcl-2
R: CTTTGAGTTTCACCGTTG (Abcam, cat# ab196495, 1:50), anti-NF-κB (Abcam, cat# ab16502, 5
Escherichia coli F: GTTAATACCTTTGCTCATTGA [45]
μg/mL) (Abcam, Cambridge, MA, USA), anti-mucin-2 (Affinity, cat#
R: ACCAGGGTATCTAATCCTGTT
All bacteria F: ACTCCTACGGGAGGCAGCAGT [45]
DF8309, 1:50) (Affinity Biotechnology Co., Ltd., Jiangsu, China) at 4 ℃
R: GTATTACCGCGGCTGCTGGCAC overnight, followed by incubation with anti-rabbit HRP (Abcam,
Cambridge, MA, USA) at 37 ℃ for 30 min. Samples were treated with a
DAB Elite kit (Beyotime Biotechnology, Shanghai, China) and photo-
Huamei Biological Co., Ltd., Wuhan, China). The level of 24 h urine graphed using optical microscopy (Olympus IX73, Shanghai, China) at
protein (24 h UP) was analyzed using an automatic clinical analyzer original magnification 100 ×, 200 × and 400 ×. The quantification of
(Hitachi High-Technologies Corporation, Tokyo, Japan). immunohistochemistry was determined by the Image-pro plus 6.0
analysis. The area of interesting (AOI) was got by selecting the stained
2.6. Intestinal microbiota analysis area on the pictures. The integrated optical density (IOD) was mea-
sured, and then followed calculation of the ratio (IOD/AOI). The mean
faeces were collected after consuming probiotics for 6 weeks and and standard deviation of each photo of the same experimental group
stored at -80 ℃. DNA was extracted by QIAamp Fast DNA stool mini kit slice were calculated, and then the relative ratio of proteins was ana-
(Qiagen, Hilden, Germany) according to the manufacturer instructions, lyzed by the Statistical Package for Social Sciences (SPSS) 11.5 soft-
and quantification was evaluated using a NanoDrop ND-1000 spectro- ware.
photometer (Thermo Scientific, Wilmington, DE). Different bacterium
in faeces samples were detected by real-time quantitative PCR, and
bacterium primers were synthesised by Dalian Bao Biotechnology Co., 2.9. Quantitative real-time PCR
Ltd, as shown in supplementary materials Table 1. Amplification and
detection of DNA were performed with a QuantiFast SYBR Green PCR Colon samples were collected and stored at -80 ℃. Total RNA was
kit (Bio-Rad, Hercules, USA). Results are expressed as the number of extracted from colon tissues using Trizol reagent (Sangon Co.,
bacteria per g of stool. Shanghai, China). The cDNA was synthesised by a First Strand cDNA
Synthesis Kit (Thermo, NY, USA). Total RNA, OligodT (15), 5×reaction
2.7. Short-chain fatty acids (SCFAs) analysis buffer, dNTPs, ribonuclease inhibitor and reverse transcriptase were
successively added into the sterile enzyme-free EP tube. Reverse tran-
The SCFAs of the feces were measured by the gas chromatography scription was performed at 42 ℃ for 60 min and then 70 ℃ for 5 min.
method according to a previous study [24]. Fresh faecal samples were PCR reaction conditions were as follows: 95 ℃ for 5 min for initial
collected and completely frozen in liquid nitrogen. Samples were denaturation; 95 ℃ for 10 s and 72 ℃ for 30 s for 40 cycles; and ex-
smashed by mortar-pestle and suspended in 1 mL of phosphoric acid 0.5 tension at 65 ℃ for 10 s, followed by dissociation curve analysis.
% per 100 mg of sample, and mixed with 2-ethylbutyradehyde added as Purified PCR products were cloned into pMD18-T and sequenced to
internal standard (IS) at a final concentration of 3 mM, diluted with verify correct amplification. Each sample was analysed in triplicate.
sterile water 1 mL, and centrifuged 10 min at 12,000 g. Supernatant was Data were calculated based on the 2−ΔΔCt method. The mRNA expres-
collected, filtered using a 0.45 μm membrane and loaded onto a gas- sion of primers is listed in supplementary materials Table 2.
phase flask for SCFAs analysis. All analyses were carried out on a gas-
phase flask (GC 2010 plus, Japan). A polar HP-INNOWAX capillary
column (30 m × 0.32 mm i.d. × 0.5 μm film thickness) (Agilent
Technologies Inc., USA) was utilized for the separation. Concentrations Table 2
Sequences of primers used in gene expression analysis.
of acetic acid, propionic acid and butyric acid (three standards) were
200 mM, 100 mM and 100 mM respectively. A volume of 1 μL of aliquot Gene Forward and reverse primers
sample was automatically injected into the inlet. Hydrogen, air, and
GPR43 F: CTGGCACAGTTCCTTGATCCTCAC
nitrogen were mixed and used as a carrier gas with the flow rates of 30
R: GGCAGCAGCAGCAACAGGAG
mL/min, 300 mL/min and 40 mL/min, respectively. The oven program GPR41 F: TGCTCATCTTCTTCGTCTGCTTCG
was adopted at an initial temperature 110 ℃ for 2.5 min, increased to R: GTCGGCTTGGAACTTGGAGGAAG
150 ℃ at a rate of 20 ℃/min, and maintained at 150 ℃ for 8 min. The GCG F: ATTCAT TGCTTGGCTGGTGA
R: CCAGAATGGTGCTCATCTCG
ratio of the peak areas of the SCFAs from the spiked and non-spiked
PC1/3 F: ACATGGGGAGAGAATCCTGTAGGCA
sample against the peak area of the internal standard is used to de- R: CATGGCCTTTGAAGGAGTTCCTTGT
termine the concentration of SCFAs. The method was validated for Claudin-1 F: CTGTGGATGTCCTGCGTTTC
linearity, limit of detection (LOD), limit of quantification (LOQ), pre- R: TCATGCACTTCATGCCAATG
cision, recovery, and stability. The IS was used to correct for injection Mucin-2 F: CCCAGAAGGGACTGTGTATG
R: TGCAGACACACTGCTCACA
variability between samples and minor changes in the instrument re-
β-actin F: GGTCATCACTATTGGCAACG
sponse. Three independent replicate extractions were performed per R: ACGGATGTCAACGTCACACT
sample with two injections each.

3
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

2.10. Western blot analysis L groups. However, there was no difference in HDL-C between the D
group and probiotic groups (P > 0.05) (Fig. 2C). Compared with the N
Pancreas samples were placed in phosphate buffered saline with group, the kidney index and 24 h UP levels in the D group were sig-
0.25 % collagenase containing Complete Mini Protease Inhibitor nificantly upregulated (P < 0.01) (Fig. 2E and F). Probiotics sig-
Cocktail (Roche, Mannheim Germany) and lysed on ice using a tissue nificantly decreased 24 h UP concentrations (P < 0.01; P < 0.05)
homogeniser. Protein was collected by centrifuging at 12,000 g for 8 compared to the D group. However, there was no difference in the
min at 4 ℃, and quantification was evaluated using the Bradford kidney index between the D and probiotic groups (P > 0.05).
method (Bio-Rad Protein Assay). A total of 50 μg of protein per sample
was separated using dodecyl sulfatepolyacrylamide gel electrophoresis 3.3. Effects of probiotics on histological changes in the pancreas, liver, and
(SDS-PAGE) and transferred onto nitrocellulose membranes (MSI, kidney
Flanders Ma) using a semi-dry transfer method. Membranes were
blocked with 5 % non-fat milk in Tris-buffered saline with Tween As shown in Fig. 3 A, pancreatic histology in the N group was
(TBST) for 2 h and incubated with primary anti-bodies in 5 % milk/ normal, with orderly arrangement of pancreatic islet cells and distinct
TBST at 4 °C overnight, including anti-Bax (Abcam, cat# ab32503, edges of islets and outer tissues. However, pancreatic histology in the D
1:1000), anti-Bcl-2 (Abcam, cat# ab196495, 1:500), anti-Caspase-3 group showed significant damage, with histomorphological features
(Abcam, cat# ab13847, 1:500), anti-AKT (Abcam, cat# ab179463, including unclear edges of the islets and outer tissues, atrophy of the
1:10000), anti-p-AKT (phospho S473) (Abcam, cat# ab81283, 1:5000), islets with inflammatory cell infiltration, and irregularly arranged
anti-PI3Kinase p85 (anti-PI3K) (Abcam, cat# ab133595, 1:1000), anti- pancreatic islet cells. Compared with the D group, both the positive and
PI3K (phospho Y607) (Abcam, cat# ab182651, 1:500), anti-interleukin probiotic groups showed significant restoration of the pancreas archi-
1β (anti-IL-1β) (Abcam, cat# ab9722, 0.1 μg/mL), anti-tumour necrosis tecture, such as reduced inflammatory cell infiltration. Liver histology
factor α (TNF-α) (Abcam, cat# ab6671, 1:500), anti-β-actin (Abcam, is shown in Fig. 3 B. Compared with the N group, abnormal histological
cat# ab8227, 1:10,000) (Abcam, Cambridge, MA, USA) and anti-in- structures in the liver from the D group showed massive diffuse lipid
terferon-γ (anti-IFN-γ, cat# BS3486, 1:500) (Bioworld Technology, Inc., macrovesicles occupying most of the hepatocyte cytoplasm. Probiotics
USA). An enhanced chemiluminescent system (Amersham Pharmacia significantly inhibited the formation of hepatic steatosis by reducing
Biotech) was applied to detect protein after incubation with horseradish lipid vesicles both in size and number. Renal histopathology results
peroxidase-conjugated antibody for 1 h. The Image J analysis was used were shown in Fig. 3 C. Renal histology in the D group showed ex-
to analyze the grayscale of the gel map and get the results of the gray cessive atrophy and fragmentation of the glomeruli, epithelial desqua-
value. The ratio of the gray value of target protein against the gray mation and degeneration. Probiotics significantly reversed abnormal
value of β-actin was the relative expression of protein. The Statistical renal histopathological changes. Overall, probiotics remarkably im-
Package for Social Sciences (SPSS) 11.5 software was used to analyze proved histological changes in the pancreas, liver and kidney.
the relative gray value of proteins
3.4. Probiotics augmented SCFA-producing bacteria and SCFAs levels
2.11. Statistical analysis
As shown in Fig. 4, Lactic acid bacteria, Bifidobacterium, Clostridium
The data were analyzed by using SPSS 11.5 software. Values were leptum, Roseburia and Prevotella levels were downregulated (P < 0.01)
presented as the means ± SD deviation. Differences among multiple (Fig. 4 A–D and F), and Escherichia coli levels in the D group were
groups were compared by one-way analysis of variance (ANOVA). significantly upregulated compared to the N group (P < 0.01) (Fig. 4
Significant difference was indicated as p < 0.05 and p < 0.01, respec- G). Conversely, probiotics decreased the number of Escherichia coli and
tively. increased the number of SCFA-producing bacteria, including Lactic acid
bacteria, Bifidobacterium, Clostridium leptum and Roseburia (P < 0.01),
3. Results which was similar to the positive control groups. In addition, there was
no significant difference in the concentration of Fusobacterium (Fig. 4
3.1. Probiotics improved FBG, OGTT, AUC, HbA1c and C-peptide levels E), Prevotella (Fig. 4 F) and total bacteria (Fig. 4 H) between the D and
probiotic groups (P > 0.05). SCFAs levels in different groups were ob-
Body weight in the D group was significantly increased, but there viously different. Compared with the N group, the levels of acetic acid,
was no difference in body weight between the D group and the pro- propionic acid and butyric acid were significantly decreased in the D
biotic groups (P > 0.05) (Fig. 1 A). Compared with the N group, FBG, group (Fig. 4 I–K) (P < 0.01). Probiotics remarkably increased the
OGTT, AUC and HbA1c levels were significantly increased (P < 0.01) concentration of propionic acid and butyric acid compared with the D
(Fig. 1 B–E), and C-peptide levels were decreased (P < 0.01) (Fig. 1 F) group (P < 0.01) (Fig. 4 J and K). Interestingly, acetic acid levels in
in the D group. The low dose of composite probiotics decreased FBG both the positive and probiotic groups were significantly decreased
after 4 week and the high dose of composite probiotics decreased FBG compared to the D group (P < 0.01) (Fig. 4 I).
after 3 week compared to the D group (P < 0.05) (Fig. 1 B). There was a
dramatic decline over time in blood glucose levels in the probiotic 3.5. Effects of probiotics on total GLP-1 and insulin level in plasma
groups (Fig. 1 C). Compared with the D group, the AUC of glucose and
HbA1c values were notably diminished (P < 0.01; P < 0.05) (Fig. 1 D To determine whether the composite probiotics can stimulate GLP-1
and E), and C-peptide levels were increased in probiotic groups secretion and promote insulin production. We evaluated the total GLP-1
(P < 0.05) (Fig. 1 F). and insulin levels of the plasma in different time after glucose feeding.
Fig. 5 A and B showed that the changes in total GLP-1 and insulin levels
3.2. Probiotics modulated TG, TC, LDL-C, HDL-C, 24 h UP levels and in plasma from 0 min to 45 min after oral glucose load. The total GLP-1
kidney index levels in the D groups were higher than that in the N group, but there
was no significant difference between the D group and the N group
Fig. 2 shows the effects of probiotics on blood lipid metabolism and (P > 0.05) (Fig. 5 A). The total GLP-1 levels in the probiotic groups
24 h UP. TG, TC and LDL-C concentrations in the D group were in- were significantly increased at 15 min compared to the D group
creased compared to the N group (P < 0.01) (Fig. 2 A, B and D). Con- (P < 0.05), but not 30 min and 45 min. The insulin levels in the D
versely, probiotics strikingly reduced TG, TC and LDL-C concentrations groups were higher than that in the N group (P < 0.05) (Fig. 5 B). The
(P < 0.01; P < 0.05), which were similar to the positive control M and insulin levels in the high-dose of probiotic group were increased at 30

4
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

Fig. 1. Effects of the composite probiotics on body weight, FBG, OGTT, AUC, HbA1c and C-peptide. (A) The body weights of mice were recorded three times a week
from 0 week to 6 week (n = 10). (B, C)The levels of FBG (n = 7) and OGTT (n = 6) were detected by the glucometer. (D) Based on the OGTT result, the AUC was
performed (n = 6). (E, F) The levels of HbA1c and C-peptide were detected by ELISA kits (n = 6). Data are presented as mean ± SD values. ## P < 0.01 vs normal
group; * P < 0.05, ** P < 0.01 vs model group. Abbreviations: FBG, Fasting blood glucose; OGTT, Oral glucose tolerance; AUC, Area under the curve.

min and 45 min compared to the D group (P < 0.05), but not 15 min. 3.6. Probiotics improved intestinal mucosal barrier function
The insulin levels in the low-dose of probiotic group were increased at
30 min compared to the D group (P < 0.05), but not 15 min and 45 min Colon histology showed a large number of goblet cells in the crypt of
(Fig. 5 B). These results showed that the composite probiotics enhanced the mucous layer, and there was no infiltration of inflammatory cells in
insulin secretion through glucose-triggered GLP-1 secretion. Ad- the laminae propria in the N group (Fig. 6 A and B). Conversely, colon
ditionally, mRNA expression of the G protein-coupled receptor 41 histological structures in the D group were crumbled, and the mucous
(GPR41), GPR43, proglucagon (GCG) and proconvertase 1/3 (PC1/3) in layer and laminae propria were infiltrated by massive inflammatory
the D group was significantly decreased compared to the N group cells, and the goblet cells of the crypt were reduced compared to the N
(P < 0.01) (Fig. 5C–F). Conversely, probiotics significantly increased group. Probiotics significantly inhibited the infiltration of inflammatory
mRNA expression of GPR41, GPR43, GCG and PC1/3 compared to the D cells and restored the number of goblet cells. The mRNA expression of
group (P < 0.01; P < 0.05). claudin-1 and mucin-2 in the D group were significantly decreased
compared to the N group (P < 0.01; P < 0.05) (Fig. 6 E and F).

Fig. 2. Effects of the composite probiotics on TG, TC, LDL-C, HDL-C, kidney index and 24 h UP. (A–D) The levels of TG, TC, LDL-C and HDL-C in the serum were
determined by ELISA kits (n = 6). The Kidney index and the 24 h UP level were detected by an automatic clinical analyzer (n = 8). Data are presented as mean ± SD
values. ## P < 0.01 vs normal group; * P < 0.05, ** P < 0.01 vs model group. Abbreviations: TC, Total cholesterol; TG, Triglyceride; LDL-C, Low density lipoprotein
cholesterol; HDL-C, High density lipoprotein cholesterol; UP, Urine protein.

5
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

Fig. 3. Effects of the composite probiotics on pancreas, liver and kidney histology. (A, B, C) Hematoxylin-eosin (HE) stained pancreatic (original magnification 200
×), hepatic (original magnification 200 ×) and renal microsections (original magnification 400 ×) (n = 4 images/group).

Compared with the D group, probiotics increased mRNA expression of immunohistochemical analysis. As shown in Fig. 7, insulin and Bcl-2
claudin-1 and mucin-2 (P < 0.01). The immunohistochemistry (IHC-P) protein expression was decreased, and Bax and NF-κB protein expres-
results of claudin-1 and mucin-2 were consistent with mRNA analysis sion was increased in the D group compared to the N group (P < 0.01).
data (Fig. 6C and D). As showed in the Fig. 6 G and H, The LPS levels of Probiotics significantly increased insulin and Bcl-2 protein expression
feces and serum in the D group were higher than that in the N group (P < 0.01), and decreased Bax and NF-κB protein expression (P < 0.01)
(P > 0.01) while probiotics significantly decreased the LPS levels in the compared to the D group (P < 0.01).
feces and serum compared to the D group (P < 0.05; P < 0.01).

3.8. Probiotics modulated the PI3K/AKT pathways


3.7. Probiotics upregulated insulin secretion by inhibiting apoptosis in the
pancreas Compared with the N group, Caspase-3, Bax, TNF-α, IL-1β and IFN-γ
protein expression was obviously increased (P < 0.01), and Bcl-2 pro-
Because insulin is a valuable hormone in regulating blood glyco- tein levels were inhibited in the D group (Fig. 8 A). Probiotics sig-
metabolism, we measured insulin secretion function in the pancreas by nificantly increased Bcl-2 protein levels and decreased Caspase-3, Bax,

Fig. 4. Effects of the composite probiotics on intestinal microbiota and SCFAs. (A–H)The levels of Lactic acid bacteria, Bifidobacterium, Clostridium leptum, Roseburia,
Fusobacterium, Prevotella, Escherichia coli and Total bacteria in the feces were determined by qRT-PCR (n = 6). (I–K) The levels of acetic acid, propionic acid and
butyric acid in the feces were determined by the gas-phase flask (n = 8). Data are presented as mean ± SD values. ## P < 0.01 vs normal group; * P < 0.05, **
P < 0.01 vs model group.

6
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

Fig. 5. The composite probiotics enhanced insulin secretion through glucose-triggered GLP-1 secretion. (A, B) The total GLP-1 and insulin levels in the plasma were
determined by ELISA kits (n = 5). (C–F) The mRNA levels of GPR43, GPR41, GCG and PC1/3 in the colon tissue were determined by qRT-PCR (n = 5). Data are
presented as mean ± SD values. # P < 0.05, ## P < 0.01 vs normal group; * P < 0.05, ** P < 0.01 vs model group. Abbreviations: GLP-1, Glucagon-like peptide-1;
GCG, Proglucagon; PC1/3, Proconvertase 1/3; GPR43/41, G protein-coupled receptor 43/41.

Fig. 6. The composite probiotics improved intestinal mucosal barrier function. (A, B) The colon histology in HE staining (original magnification 100 × and 200 ×)
(n = 4 images/group). (C, D) The claudin-1 (original magnification 200 ×) and mucin-2 (original magnification 400 ×) protein expression of the colon tissue in
immunohistochemistry staining (n = 4 images/group). (E, F) The mRNA levels of claudin-1 and mucin-2 in the colon tissue were determined by qRT-PCR (n = 5).
(G, H) The levels of LPS in the faeces and serum were determined by ELISA kits (n = 6). Mean integral optical density values of claudin-1 and mucin-2 were analyzed
by Image-Pro Plus 6.0. Data are presented as mean ± SD values. ## P < 0.01 vs normal group; * P < 0.05, ** P < 0.01 vs model group.

TNF-α, IL-1β and IFN-γ protein levels compared to the D group 4. Discussion
(P < 0.01). The PI3K/AKT signalling pathway plays an important role
in regulating insulin secretion in the pancreas. As shown in Fig. 8B, Probiotics have many functions, including anti-oxidation, anti-
protein expression of p-PI3K/t-p-PI3K and p-AKT/t-AKT in the D group cancer, anti-inflammation, and improved metabolism and im-
was significantly decreased compared to the N group (P < 0.01; munological function [28,25,26]. An increasing number of studies have
P < 0.05). Compared with the D group, probiotics significantly upre- shown that probiotics have enormous potential in treating metabolic
gulated p-PI3K/t-p-PI3K and p-AKT/t-AKT protein levels (P < 0.01). diseases, such as diabetes and obesity [27,28]. Hsieh et al. reported that
Lactobacillus reuteri GMNL-263 improved insulin resistance by reg-
ulating T2D-related parameters in high fructose-fed rats, such as

7
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

Fig. 7. Effects of the composite probiotics on the protein expression of insulin, Bax, Bcl-2 and NF-κB in the pancreas. (A–D) The protein levels of the insulin (original
magnification 200 ×), Bax (original magnification 400 ×), Bcl-2 (original magnification 400 ×) and NF-κB (original magnification 200 ×) in im-
munohistochemistry staining (n = 4 images/group). Mean integral optical density values of insulin, Bax, Bcl-2 and NF-κB were analyzed by Image-Pro Plus 6.0. Data
are presented as mean ± SD values. ## P < 0.01 vs normal group; ** P < 0.01 vs model group.

OGTTs, glycated haemoglobin and insulin [29]. In a similar study, showed that oral administration of 14 composite probiotics sig-
Balakumar et al. showed that a combination of Lactobacillus plantarum nificantly improved blood glucose and blood lipid related parameters,
and Lactobacillus fermentum improved insulin sensitivity and OGTTs in including FBG, OGTT, HbA1c, C-peptide, TG, TC and LDL-C (Figs. 1 and
C57BL/6 J mice fed a high fat diet [30]. The leptin receptor mutation of 2).
the 4 chromosome in the C57BL/Ks strain mice causes obesity, hy- The representative characteristics of intestinal microbiota in T2D
perglycemia and islet resistant, which is very similar to the pathogen- patients include a decrease in the number of SCFA-producing bacteria
esis of human T2D [31,32]. Therefore, we used the C57BL/Ks mice as and an increase in harmful bacteria. Okouchi et al. found that the levels
the normal group and the db/db mice as the diabetic mice for exploring of Clostridiaceae, Bifidobacterium, Lactobacillus and Escherichia in the
the possible anti-diabetic mechanisms of 14 probiotics. Our results normal diet mice were higher than other bacterial [33]. Some studies in

Fig. 8. The composite probiotics protected pancreas against apoptosis by upregulation PI3K/AKT. (A) The protein levels of Caspase-3, Bax, Bcl-2, TNF-α, IFN-γ and
IL-1β were detected by the Wester-blot (n = 5). (B) The protein levels of p-PI3K/t-PI3K and p-AKT/t-AKT were detected by the Wester-blot (n = 5). The gel map and
the gray value were analyzed by Image J. Data are presented as mean ± SD values. # P < 0.05, ## P < 0.01 vs normal group; * P < 0.05, ** P < 0.01 vs model group.
Abbreviations: TNF-α, Tumor necrosis factor α; IFN-γ, Interferon-γ; IL-1β, Interleukin-1β; PI3K/AKT, Phosphatidylinositol 3ʹ-kinase/protein kinase B; Bcl-2, B-cell
lymphoma 2; Bax, Bcl-2-associated X protein.

8
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

human reported that Prevotella, Roseburia, Eubacterium halii, Bacteroides the GCG mRNA level of intestine tissue in the leptin-resistant 2 diabetic
fragilis and Faecalibacterium prauznitzii concentrations were decreased model mice (db/db mice) was higher than the normal group (C57BL/
and that Lactobacillus gasseri, Streptococcus mutans and Escherichia coli KsJ mice) [52]. Lu et al. have a similar report that the GCG mRNA level
were increased in T2D patients [34,35]. Several animal studies have of the ileum in the STZ-induced diabetic rat (Sprague-Dawley rat) was
shown that the number of Lactobacillus, Bifidobacterium, Clostridium higher than the normal group, but there was no difference [53]. Mor-
leptum, Bacteroides and Prevotella was significantly decreased in a T2D imoto et al. reported that there is no difference in the GCG mRNA levels
rat model induced by a high fat diet and streptozotocin injection of the terminal ileum between the C57BL/6 J mice receiving high-fat
[35,36]. Previous studies have shown that probiotics remarkably in- diet and the C57BL/6 J mice receiving the normal diet [54]. Our resluts
creased the levels of these SCFA-producing bacteria and decreased the showed that the GCG and PC1/3 mRNA levels were significantly de-
levels of Escherichia [26]. The SCFA-producing bacteria benefit to the creased in the D group compared to the N group while probiotics in-
host by inhibiting the level of harmful bacterial through struggling for creased GCG and PC1/3 mRNA levels. The changes in the GCG mRNA
the limited nutrients and living space, preventing the pathogen invasion expression are strikingly different might due to diversified influential
from mucosa and mitigating inflammation [37]. In the study, 14 pro- factors, including the pathophysiological characteristics of animal
biotics significantly decreased Escherichia coli levels and increased the model, the examination of tissue location, feeding time and various
number of SCFA-producing bacteria in db/db mice (Fig. 4). In fact, experimental conditions.
changes in intestinal microbiota are strikingly different in different The pancreas is an important organ in regulating glycometabolism
individuals due to diversified influential factors, including genetics, because it is the only tissue that secretes insulin. High expression of
diet, species and more [38]. Another reason for differences in intestinal tumour necrosis factor-α (TNF-α), interleukin-1β, and interferon-γ in-
microbiota is that research subjects may be different and studies are duced β-cell apoptosis and dysfunction [55]. β-cell apoptosis in strep-
typically performed under various experimental conditions [39]. tozocin-induced diabetic rats was inhibited by inactivating NF-κB ac-
SCFAs play an important role in postponing T2D progression by tivity, which improved β-cell function and morphology [56].
imprving the function of the intestinal mucosa barrier. SCFAs can Accumulating evidence has demonstrated that the PI3K/AKT cascade is
supply energy for enterocytes and reduce production of toxic substances essential for ameliorating insulin sensitivity and protecting β-cells
and inflammation by inhibiting the growth of harmful bacteria [40]. against apoptosis by regulating the expression of Bcl-2 and Bax [57].
Additionally, SCFAs can repaire and improve the function of intestinal Cheng et al. found that LY294002, a PI3K/AKT inhibitor, obviously
mucosa barrier by pormoting the synthesis of intestinal mucosin and reduced anti-apoptotic effects on Min6 cells by blocking PI3K/AKT
mucus secretion of intestinal epithelial cells, inhibting the pro-in- activation [58]. These reports are consistent with the results of the
flammatory factors secretion, increasing anti-inflammatory IL-10 pro- present study, 14 probiotics significantly reduced protein expression of
duction and activing Treg cell activity [41,42]. Some studies reported inflammatory factors TNF-α, IL-1β and IFN-γ, inhibited pro-apoptotic
that SCFAs induced by Lactobacillus strengthened the function of the protein expression (Bax and caspase-3) and increased anti-apoptotic
intestinal mucosa barrier by upregulating tight junction protein ex- Bcl-2, PI3K/AKT protein expression (Fig. 8), suggesting that 14 pro-
pression and inhibiting lipopolysaccharide (LPS) or endotoxin infiltra- biotics might improve β-cell function and protect β-cells against
tion, thus upregulating insulin sensitivity [43]. It is reported that LPS apoptosis by upregulating PI3K/AKT activation. Notably, Hansen et al.
mainly from the Gram-negative bacteria (Escherichia coli) can enter the found that prebiotics could portect against autoimmune destruction of
bloodstream through the leaky gut, then inducing the insulin-intensive β cells by regulating T cell activity and improving the gut barrier fuc-
organs (pancreas, liver and kidney) inflammation [36,44]. Thus the tion in NOD mice with autoimmune diabetes, but not the regulation of
serum LPS levels can indirectly reflect the intestinal permeability. These glucose-insulin and the production of GLP-1 and SCFAs [59]. Dolpady
reports are consistent with our results in that probiotics improved the et al. had a simlilar study that Lactobacillaceae-enriched probiotic
gut barrier fuction by increasing SCFAs levels, upregulating claudin-1 VSL#3 could alleviate T1D by modulating the microbiota composition
and mucin-2 expression and downregulating LPS levels (Figs. 4 and 6). and Teff/Treg cell balance and restoring gut immunue homeostasis
In the intestine, GPR43 and GPR41 benefit to regulate the nenegy [60]. These studies suggest that probiotics and prebiotics might have
homeostasis by affecting the incretion hormones secretion and nutrients differences in improvement of GLP-1 secretion depened on different
absorption. It is reported that the SCFAs can active the G protein-cou- signal pathways, which might be related with the development and
pled receptors (GPR43/41) expressed by L cells in the colon [45,46]. pathophysiological processes of T1D and T2D [61].
GPR43 and GPR41 were SCFA receptors that respectively comple with The study may aid the understanding of the mechanisms underling
Gq- or Gi/o family G proteins and Gi-family G proteins [47]. Acetate, 14 probiotics treatment for T2D. However, there are still many ob-
propionate and butyrate are important metabolic production of in- stacles in the clinical applications of probiotics in T2D therapy. First,
testinal microbiota. GPR41 is sensitive to propionate and butyrate while there are differences in the stability and environment of gut microbiota
GPR43 is more sensitive to acetate and propionate than to butyrate due to the species and individual differences between rodents and hu-
[48]. Some studies have reported that these SCFA-producing bacteria mans [62]. Second, the appropriate dosages and the colonization sta-
(mainly Lactobacillus and Bifidobacterium) can promote the GLP-1 se- bility of these probiotics in human intestinal tract are not clear. Third,
cretion by upregulating GPR43/41 activity in the intestine, followed by there are multifarious metabolites produced by the host’ microbiota are
GLP-1-induced insulin secretion, which effectively adjusts and controls involved in the regulation of metabolic pathways, immune-in-
blood glucose metabolism [49,50]. In the study, we found that the flammatory pathways and so on [63]. These intricate regulatory me-
number of these SCFA-producing bacteria was increased by the com- chanisms are not entirely clear. Dispite theses challenges, It's still worth
posite probiotics. Additionally, levels of propionic acid and butyric acid trying to unlock the secrets of probiotics to host, which has important
were significantly increased in the composite probiotic groups as well therapeutic potential for metabolic diseases and other disease.
as the upregulation activity of GPR43 and GPR41. These results in-
dicated that 14 probiotics promoted GLP-1 secretion might be depen- 5. Conclusion
dent on the upregultaion of GPR43/41 activity. In addition, GLP-1 can
be produced by the pathway in which proglucagon (GCG) is cleaved by These probiotics effectively improved blood glucose and blood lipid
the enzyme proconvertase 1/3 (PC1/3). Rohden et al. found that lower parameters so as to delay the development of T2D. Probiotics improved
PC1/3 gene expression can attenuate bioactivation of pro-glucose de- the gut barrier function through increasing the levels of SCFA-produ-
pendent insulinotropic peptide and proglucagon in T2D patients [51]. cing bacteria and SCFAs as well as the expression of cluaudin-1 and
Liu et al. found that the CGC mRNA level of intestine tissue in STZ- mucin-2, and decreasing Escherichia coli and LPS levels. Probiotics sig-
induced ICR diabetic mice was lower than the normal group. However, nificantly promoted GLP-1 secretion by upregulating the activities of

9
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

GPR43/41, GCG and PC1/3. Furthermore, probiotics improved pan- [23] M. Le Barz, F.F. Anhê, T.V. Varin, et al., Probiotics as complementary treatment for
creatic function and inhibited apoptosis in the pancreas by upregulating metabolic disorders, Diabetes Metab. J. 39 (2015) 291–303.
[24] L.L. Wang, H.H. Guo, S. Huang, et al., Comprehensive evaluation of SCFA pro-
PI3K/AKT activation. These results indicated that 14 composite pro- duction in the intestinal bacteria regulated by berberine using gas-chromatography
biotics might be considered to be a potential treatment option for combined with polymerase chain reaction, J. Chromatogr. B Analyt. Technol.
treating patients with T2D. Biomed. Life Sci. 2015 (2017) 70–80.
[25] J.Y. Yoo, S.S. Kim, Probiotics and prebiotics: present status and future perspectives
on metabolic disorders, Nutrients 8 (2016) 173.
Declaration of Competing Interest [26] S. Singh, R.K. Sharma, S. Malhotra, et al., Lactobacillus rhamnosus NCDC17 ame-
liorates type-2 diabetes by improving gut function, oxidative stress and in-
flammation in high-fat-diet fed and streptozotocintreated rats, Benef. Microbes 8
No competing interests are associated with study. (2017) 243–255.
[27] M.C. Mekkes, T.C. Weenen, R.J. Brummer, et al., The development of probiotic
Acknowledgments treatment in obesity: a review, Benef. Microbes 5 (2014) 19–28.
[28] A.N. Payne, C. Chassard, M. Zimmermann, et al., The metabolic activity of gut
microbiota in obese children is increased compared with normal-weight children
This project was supported by the funds from University Scientific and exhibits more exhaustive substrate utilization, Nutr. Diabetes 18 (2011) e12.
Research Plan of Xinjiang Uygur Autonomous Region, China [29] F.C. Hsieh, C.L. Lee, C.Y. Chai, et al., Oral administration of Lactobacillus reuteri
(XJEDU2020I012) and Graduate Innovation and Entrepreneurship GMNL-263 improves insulin resistance and ameliorates hepatic steatosis in high
fructose-fed rats, Nutr. Metab (Lond). 10 (2013) 35.
Project of Xinjiang Uygur Autonomous Region, China (XJ2019G177). [30] M. Balakumar, D. Prabhu, C. Sathishkumar, et al., Improvement in glucose toler-
ance and insulin sensitivity by probiotic strains of Indian gut origin in high-fat diet-
References fed C57BL/6J mice, Eur. J. Nutr. 57 (2018) 279–295.
[31] M.S. Islam, Animal models of diabetic neuropathy: progress since 1960s, J. Diabetes
Res. 2013 (2013) 149452.
[1] R. Tojo, A. Suárez, M.G. Clemente, et al., Intestinal microbiota in health and dis- [32] K.P. Hummel, M.M. Dickie, D.L. Coleman, Diabetes, a new mutation in the mouse,
ease: role of bifidobacteria in gut homeostasis, World J. Gastroenterol. 20 (2014) Science 153 (1966) 1127–1128.
15163–15176. [33] R. Okouchi, S.E. Yamamoto K.et al., Simultaneous intake of Euglena gracilis and
[2] J.E. Shaw, R.A. Sicree, P.Z. Zimmet, Global estimates of the prevalence of diabetes vegetables exerts synergistic anti-obesity and anti-inflammatory effects by mod-
for 2010 and 2030, Diabetes Res. Clin. Pract. 87 (2010) 4–14. ulating the gut microbiota in diet-induced obese mice, Nutrients 11 (2019).
[3] Y.S. Lee, H.S. Jun, Anti-diabetic actions of glucagon-like peptide-1 on pancreatic [34] M. Sedighi, S. Razavi, F. Navab-Moghadam, et al., Comparison of gut microbiota in
beta-cells, Metabolism 63 (2014) 9–19. adult patients with type 2 diabetes and healthy individuals, Microb. Pathog. 111
[4] T.D. Müller, C. Clemmensen, B. Finan, et al., Glucagon-like peptide 1 (GLP-1), (2017) 362–369.
Pharmacol. Rev. 70 (2018) 712–746. [35] A. Muñoz-Garach, C. Diaz-Perdigones, F.J. Tinahones, Gut microbiota and type 2
[5] K. Timper, E. Dalmas, E. Dror, et al., Glucose-dependent insulinotropic peptide diabetes mellitus, Endocrinol. Nutr. 63 (2016) 560–568.
stimulates glucagon-like peptide 1 production by pancreatic islets via interleukin 6, [36] K.K. Li, P.J. Tian, S.D. Wang, et al., Targeting gut microbiota: lactobacillus alle-
produced by a cells, Gastroenterology 151 (2016) 165–179. viated type 2 diabetes via inhibiting LPS secretion and activating GPR43 pathway,
[6] L. Wang, P. Li, Z. Tang, et al., Structural modulation of the gut microbiota and the J. Funct. Foods 38 (2017) 561–570.
relationship with body weight: compared evaluation of liraglutide and saxagliptin [37] K.M. Maslowski, A.T. Vieira, A. Ng, et al., Regulation of inflammatory responses by
treatment, Sci. Rep. 6 (2016) 33251. gut microbiota and chemoattractant receptor GPR43, Nature 461 (2009)
[7] J. Zhang, H. Su, Q. Li, et al., Oral administration of Clostridium butyricum 1282–1286.
CGMCC0313-1 inhibits beta-lactoglobulin-induced intestinal anaphylaxis in a [38] M. Rastelli, C. Knauf, P.D. Cani, Gut Microbes and Health: A Focus on the
mouse model of food allergy, Gut Pathog. 9 (2017) 11. Mechanisms Linking Microbes, Obesity, and Related Disorders, Obesity Silver
[8] F. Hendijani, V. Akbari, Probiotic supplementation for management of cardiovas- Spring (Silver Spring) 26 (2018) 792–800.
cular risk factors in adults with type II diabetes: a systematic review and meta- [39] J. Miyamoto, K. Watanabe, S. Taira, et al., Barley beta-glucan improves metabolic
analysis, Clin. Nutr. 37 (2018) 532–541. condition via short-chain fatty acids produced by gut microbial fermentation in
[9] J. Qin, Y. Li, Z. Cai, et al., A metagenome-wide association study of gut microbiota high fat diet fed mice, PLoS One 13 (2018) e0196579.
in type 2 diabetes, Nature 490 (2012) 55–60. [40] Q. Zhang, X. Xiao, M. Li, et al., Vildagliptin increases butyrate-producing bacteria in
[10] J.F. Garcia-Mazcorro, N.N. Lage, S. Mertens-Talcott, et al., Effect of dark sweet the gut of diabetic rats, PLoS One 12 (2017) e0184735.
cherry powder consumption on the gut microbiota, short-chain fatty acids, and [41] W. Miao, X. Wu, K. Wang, et al., Sodium butyrate protes reassembly of tight
biomarkers of gut health in obese db/db mice, Peer J. 6 (2018) e4195. junctions in Caco-2 monolayers InvolvingInhibition of MLCK/MLC2 pathway and
[11] L. Wang, Y. Tao, Z. Xie, et al., The metabolic syndrome in Uygur and Kazak po- phosphorylation of PKCβ2, Int. J. Mol. Sci. 17 (2016).
pulations/Prevalence of metabolic syndrome, insulin resistance, impaired fasting [42] B. Shen, J. Hu, H. Song, et al., Antibiotics exacerbated colitis by affecting the mi-
blood glucose,and dyslipidemia in Uygur and Kazak populations, J. Clin. Hypertens. crobiota, Treg cells and SCFAs in IL10-deficient mice, Biomed. Pharmacother. 114
Greenwich (Greenwich) 12 (2010) 741–745. (2019) 108849.
[12] N. Li, H. Wang, Z. Yan, et al., Ethnic disparities in the clustering of risk factors for [43] S. Seo, J.S. Shin, W.S. Lee, et al., Anti-colitis effect of Lactobacillus sakei K040706
cardiovascular disease among the Kazakh, Uygur, Mongolian and Han populations via suppression of inflammatory responses in the dextran sulfate sodium-induced
of Xinjiang: a cross-sectional study, BMC Public Health 12 (2012) 499. colitis mice model, J. Funct. Foods 29 (2017) 256–268.
[13] W.L. Yan, X.S. Li, Q. Wang, et al., Overweight, high blood pressure and impaired [44] J. Xu, R. Liang, W. Zhang, et al., Faecalibacterium prausnitzii-derived microbial
fasting glucose in Uyghur, Han, and Kazakh Chinese children and adolescents, Ethn. anti-inflammatory molecule regulates intestinal integrity in diabetes mellitus mice
Health 20 (2015) 365–375. via modulating tight junction protein expression, J. Diabetes (2019).
[14] T. Manaer, L. Yu, Y. Zhang, et al., Anti-diabetic effects of shubat in type 2 diabetic [45] W. Wang, L. Chen, R. Zhou, et al., Increased proportions of Bifidobacterium and the
rats induced by combination of high-glucose-fat diet and low-dose streptozotocin, J. Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel
Ethnopharmacol. 169 (2015) 269–274. disease, J. Clin. Microbiol. 52 (2014) 398–406.
[15] R.A. Bagarolli, N. Tobar, A.G. Oliveira, et al., Probiotics modulate gut microbiota [46] B.S. Samuel, A. Shaito, T. Motoike, et al., Effects of the gut microbiota on host
and improve insulin sensitivity in DIO mice, J. Nutr. Biochem. 50 (2017) 16–25. adiposity are modulated by the short-chain fatty-acid binding G protein-coupled
[16] D. Dinareer, L. Lu, S. Jialehasibieke, et al., Probiotic characteristics of the lactic acid receptor, Gpr41, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 16767–16772.
bacteria and yeasts in Xinjiang-traditional fermented dairy products, China [47] I. Kimura, A. Ichimura, R. Ohue-kitano, et al., Free fatty acid receptors in health and
Microecology J. 30 (2018) 5-9+13. disease, Physiol. Rev. 100 (2020) 171–210.
[17] S. Jialehasibieke, S. Xin, J. Amanguli, et al., Probiotic characteristics of probiotics in [48] J. Miyamoto, R. Ohue-Kitano, H. Mukouyama, et al., Ketone body receptor GPR43
Xinjiang traditional fermented dairy products, China Microecology J. 31 (2019) regulates lipid metabolism under ketogenic conditions, Proc. Natl. Acad. Sci. U. S.
502–508. A. 116 (2019) 23813–23821.
[18] H.A. Lee, H. Kim, W.K. Lee, et al., Dead Lactobacillus plantarum stimulates and [49] R. Aoki, K. Kamikado, W. Suda, et al., A proliferative probiotic Bifidobacterium
skews immune responses toward t helper 1 and 17 polarizations in RAW 264.7 cells strain in the gut ameliorates progression of metabolic disorders via microbiota
and mouse splenocytes, J. Microbiol. Biotechnol. 26 (2016) 469–476. modulation and acetate elevation, Sci. Rep. 7 (2017) 43522.
[19] L.S. Dalbøge, D.L. Almholt, T.S. Neerup, et al., Characterisation of age-dependent [50] Y. Liang, S. Liang, Y. Zhang, et al., Oral administration of compound probiotics
beta cell dynamics in the male db/db mice, PLoS One 8 (2013) e82813. ameliorates HFD-Induced gut microbe dysbiosis and chronic metabolic inflamma-
[20] K.H. Kim, I.S. Lee, J.Y. Park, et al., Cucurbitacin B induces hypoglycemic effect in tion via the g protein-coupled receptor 43 in non-alcoholic fatty liver disease rats,
diabetic mice by regulation of AMP-Activated protein kinase alpha and glucagon- probiotics antimicrob, Proteins 11 (2019) 175–185.
like Peptide-1 via bitter t aste receptor signaling, Front. Pharmacol. 9 (2018) 1071. [51] F. Rohden, C.S. Costa, T.O. Hammes, et al., Obesity associated with type 2 diabetes
[21] P. Chen, L. Li, X. Xu, et al., Liraglutide improved inflammation via mediating IL-23/ mellitus is linked to decreased PC1/3 mRNA expression in the Jejunum, Obes. Surg.
Th-17 pathway in obese diabetic mice with psoriasiform skin, J. Dermatolog. Treat. 24 (2014) 2075–2081.
23 (2019) 1–21. [52] Y.X. Liu, M.M. Si, W. Lu, et al., Effects and molecular mechanisms of the anti-
[22] S.E. Jang, M.J. Han, S.Y. Kim, et al., Lactobacillus plantarum CLP-0611 ameliorates diabetic fraction of Acorus calamus L. On GLP-1 expression and secretion in vivo
colitis in mice by polarizing M1 to M2-like macrophages, Int. Immunopharmacol. and in vitro, J. Ethnopharmacol. 166 (2015) 168–175.
21 (2014) 186–192. [53] S.S. Lu, Y.L. Yu, H.J. Zhu, et al., Berberine promotes glucagon-like peptide-1 (7–36)

10
Y. Wang, et al. Biomedicine & Pharmacotherapy 125 (2020) 109914

amide secretion in streptozotocin-induced diabetic rats, J. Endocrinol. 200 (2009) [59] C.H.F. Hansen, D.S. Larsen, H.O. Petersson, et al., Targeting gut microbiota and
159–165. barrier function with prebiotics to alleviate autoimmune manifestations in NOD
[54] K. Morimoto, M. Watanabe, T. Sugizaki, et al., Intestinal bile acid composition mice, Diabetologia 62 (2019) 1689–1700.
modulates prohormone convertase 1/3 (PC1/3) expression and consequent GLP-1 [60] J. Dolpady, C. Sorini, C. Di Pietro, et al., Oral probiotic VSL#3 prevents auto-
production in male mice, Endocrinology 157 (2016) 1071–1081. immune diabetes by modulating microbiota and promoting indoleamine 2,3-
[55] W. You, K. Wang, C. Yu, et al., Baicalin prevents tumor necrosis factor-alpha-in- Dioxygenase-Enriched tolerogenic intestinal environment, J. Diabetes Res. 2016
duced apoptosis and dysfunction of pancreatic beta-cell line Min6 via upregulation (2016) 7569431.
of miR-205, J. Cell. Biochem. 119 (2018) 8547–8554. [61] T.J. O’Malley, G.E. Fava, Y. Zhang, et al., Progressive change of intra-islet GLP-1
[56] V. Kondylis, S. Kumari, K. Vlantis, et al., The interplay of IKK, NF-kappaB and production during diabetes development, Diabetes Metab. Res. Rev. 30 (2014)
RIPK1 signaling in the regulation of cell death, tissue homeostasis and inflamma- 661–668.
tion, Immunol. Rev. 227 (2017) 113–127. [62] M.E. Sanders, D.J. Mernstein, G. Reid, et al., Probiotics and prebiotics in intestinal
[57] T.L. Jetton, J. Lausier, K. LaRock, et al., Mechanisms of compensatory beta-cell health and disease: from biology to the clinic, Nat. Rev. Gastroenterol. Hepatol. 16
growth in insulin-resistant rats: roles of Akt kinase, Diabetes 54 (2005) 2294–2304. (2019) 605–616.
[58] Q. Cheng, W. Dong, L. Qian, et al., Visfatin inhibits apoptosis of pancreatic beta-cell [63] D. Kothari, S. Patel, S.K. Kim, Probiotic supplements might not be universally-ef-
line, MIN6, via the mitogen-activated protein kinase/phosphoinositide 3-kinase fective and safe: a review, Biomed. Pharmacother. 111 (2019) 537–547.
pathway, J. Mol. Endocrinol. 47 (2011) 13–21.

11

You might also like