Skip to main content
Erschienen in: Journal of Translational Medicine 1/2023

Open Access 01.12.2023 | Review

The effects of probiotics supplementation on glycaemic control among adults with type 2 diabetes mellitus: a systematic review and meta-analysis of randomised clinical trials

verfasst von: Guang Li, Hao Feng, Xin-Liang Mao, Yan-Jun Deng, Xiao-Bao Wang, Qiong Zhang, Yan Guo, Su-Mei Xiao

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2023

Abstract

Objective

This systematic review and meta-analysis study aimed to evaluate the effectiveness of probiotics supplementation on glycaemic control in patients with type 2 diabetes mellitus (T2DM) based on the data from the randomised clinical trials (RCTs).

Methods

PubMed, Web of Sciences, Embase, and Cochrane Library were searched from the inception to October 2022, and RCTs about probiotics and T2DM were collected. The standardised mean difference (SMD) with 95% confidence interval (CI) was used to estimate the effects of probiotics supplementation on glycaemic control related parameters, e.g. fasting blood glucose (FBG), insulin, haemoglobin A1c (HbA1c), and homeostasis model of assessment of insulin resistance (HOMA-IR).

Results

Thirty RCTs including 1,827 T2MD patients were identified. Compared with the placebo group, the probiotics supplementation group had a significant decrease in the parameters of glycaemic control, including FBG (SMD = − 0.331, 95% CI  − 0.424 to − 0.238, Peffect < 0.001), insulin (SMD = − 0.185, 95% CI  − 0.313 to − 0.056, Peffect = 0.005), HbA1c (SMD = − 0.421, 95% CI  − 0.584 to − 0.258, Peffect < 0.001), and HOMA-IR (SMD = − 0.224, 95% CI  − 0.342 to − 0.105, Peffect < 0.001). Further subgroup analyses showed that the effect was larger in the subgroups of Caucasians, high baseline body mass index (BMI ≥ 30.0 kg/m2), Bifidobacterium and food-type probiotics (Psubgroup < 0.050).

Conclusion

This study supported that probiotics supplementation had favourable effects on glycaemic control in T2DM patients. It may be a promising adjuvant therapy for patients with T2DM.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-023-04306-0.
Guang Li, Hao Feng and Xin-Liang Mao are contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
T2DM
Type 2 diabetes mellitus
SMD
Standardised mean difference
FBG
Fasting blood glucose
HbA1c
Haemoglobin A1c
HOMA-IR
Homeostasis model of assessment of insulin resistance
RCTs
Randomised controlled trials
BMI
Body mass index
SD
Standard deviation
SE
Standard error
CI
Confidence interval
CFU
Colony-forming units
IBS
Irritable bowel syndrome

Introduction

Type 2 diabetes mellitus (T2DM), an endocrine and metabolic disease, is influenced by host physiology and environmental factors [1]. More than 500 million people are living with diabetes globally, and this number is expected to increase to 783 million by 2045 [2]. T2DM is a common disease that accounts for approximately 90% of all cases of diabetes [3], and it may cause reduced life expectancy and life-threatening and costly complications [4]. There is no radical cure for T2DM [5, 6], and its treatment relies on the long-term use of anti-diabetic drugs [7, 8]. Therefore, it is crucial to explore new methods that may effectively delay or even reverse the progression of T2DM.
Recent studies have shown that the gut microbiota plays a key role in the maintenance of host homeostasis and pathogenesis of T2DM [9, 10]. Probiotics are microbial dietary supplements that alter the gut microbiota. Some randomised controlled trials (RCTs) have investigated the effects of probiotic interventions on glycaemic control in T2DM patients. However, evidence from clinical trials on the effects of probiotic supplementation on glycaemic control remains inconsistent. Asemi et al. [11] conducted a randomised double-blind placebo-controlled clinical trial involving 54 T2DM patients, which revealed that multi-species probiotic (mixture of Lactobacillus and Bifidobacterium) supplementation prevented an increase in the fasting blood glucose (FBG) level from baseline in these patients. Meanwhile, Razmpoosh et al. [12] randomly assigned 60 T2DM patients into two groups to take either a probiotic (mixture of Lactobacillus and Bifidobacterium) or a placebo intervention, and the results showed no significant differences in insulin or insulin resistance levels between the two groups. In 2016, Li et al. performed a systematic review and meta-analysis of 12 RCTs with 714 individuals and reported that probiotic supplementation could alleviate FBG, but no significant differences were observed in the haemoglobin A1c (HbA1c) level or homeostatic model assessment of insulin resistance (HOMA-IR) score between the probiotic and control groups of T2DM patients [13]. In 2020, Tao et al. systematically summarised 15 RCTs with 902 individuals, and the results of the meta-analysis indicated that probiotic supplementation reduced HbA1c, FBG and insulin resistance levels in T2DM patients [14]. However, some related RCTs (n = 11, including 630 patients) were not included in their study. Since then, more RCTs (n = 6) of the effects of probiotic supplementation on glycaemic control, including a total of 511 T2DM patients, have been reported [15, 16]. Controversy still exists regarding the effects of probiotics on glycaemic control in T2DM patients. Variations in participant (e.g. race) and intervention characteristics (e.g. dose, probiotic genus, and duration) in different studies may have given rise to the contradictory results. No study has detected differences in the effects of probiotic supplementation on glycaemic control according to the participant and intervention characteristics.
In this systematic review and meta-analysis, we aimed to evaluate the effects of a probiotic intervention on glycaemic control in T2DM patients and to evaluate the variations in these effects due to participant characteristics, e.g. race and baseline body mass index (BMI), and intervention characteristics, e.g. the probiotic dose, the duration of the intervention, the probiotic genus, and the type of vehicle used to deliver the probiotics.

Methods

This study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement [17] (Additional file 1: Table S1). The protocol for this study has been registered at the International Prospective Register of Systematic Reviews (registration number: CRD42022370226).

Search strategy

Two reviewers (Guang Li and Yan-Jun Deng) independently searched PubMed, Web of Science, Embase, and Cochrane Library databases from their inception until October 2022 using various probiotic-related words and Medical Subject Heading terms in combination with ‘T2DM’ (Additional file 2: Table S2). No language or other restrictions were applied during the search, and all relevant studies were found to be published in English. A manual search was also performed to identify relevant studies from the references of the included studies.

Inclusion and exclusion criteria

Studies were included in the analysis if: (1) the participants were T2DM patients aged ≥ 18 years; (2) the study design was an RCT; (3) the intervention was the intake of probiotics from supplements and/or food; (4) the control group received a placebo intervention; and (5) the main outcomes included the glycaemic profile, e.g. FBG, insulin, and HbA1c levels and the HOMA-IR score. Studies were excluded from the analysis if: (1) the participants had other types of diabetes, e.g. gestational diabetes or type 1 diabetes or (2) the participants were concurrently receiving other interventions, e.g. synbiotics, herbs, prebiotics, or micro- nutrients.

Data extraction and quality assessment

Two researchers (Guang Li and Yan-Jun Deng) independently performed the literature search and data extraction, and disagreements were resolved by a third senior researcher (Su-Mei Xiao). Basic information (e.g. first author, year, and country of the study and the age, sex, and BMI of the participants), the study design, intervention information (probiotic genus and dose and duration of the intervention), and outcomes were extracted from the included studies. Two reviewers (Xiao-Bao Wang and Qiong Zhang) evaluated the quality of the included studies using the Cochrane risk-of-bias assessment tool. The risk of bias in the included studies was classified as low, unclear, or high.

Data synthesis and statistical analysis

The change in glycaemic control parameters was the primary outcome in this study. It was calculated as the final measurement value minus the baseline measurement value in each group. The mean and standard deviation (SD) of the change in glycaemic control parameters for the control group and the intervention group were extracted, respectively. If the study provided the standard error (SE) of mean change, the SE was converted to SD based on the sample size. For studies that did not directly report SD of mean change, the SDs of the baseline and final measurement values and the correlation coefficient (Corr) were used to calculated SDEffect,change (SDE,change) according to the following formula [18]:
$${\mathrm{SD}}_{\mathrm{E},\mathrm{change}}=\sqrt{{SD}_{E,baseline}^{2}+{SD}_{E,final}^{2}-(2 * Corr * {SD}_{E,baseline} * {SD}_{E, final})}$$
Corr is the correlation coefficient between the baseline and final measurement values. For the pretest–posttest design, presumably the correlation is at least 0.5. This was the Corr estimate value being used to impute the missing SDs of mean change in this study [18, 19]. If the study presented data in medians and quartiles, the mean and SD values were estimated [20, 21]. If the intervention included multiple time points, the longest intervention time was included in the analysis.
The standardised mean difference (SMD) with the 95% confidence interval (CI) was used to assess the effects of probiotic interventions on glycaemic control in T2DM patients. The boundary values of the SMD were set at 0.2, 0.5, and 0.8, corresponding to small, medium, and large effects, respectively [22]. Heterogeneity was assessed using Cochrane’s Q statistic (chi-square). The inverse variance (I2) was used to assess the size of the heterogeneity. A fixed-effects model was used for the meta-analysis when I2 ≤ 50%, and a random-effects model was used when I2 ≥ 50%. Subgroup analysis was used to explore the possible sources of heterogeneity. Subgroup analyses were performed for race (Asian vs. Caucasian), probiotic dose (≤ 1 × 1010 colony-forming units (CFU)/day vs. > 1 × 1010 CFU/day), the duration of the intervention (≤ 8 weeks vs. > 8 weeks), probiotic genus (Lactobacillus, Bifidobacterium, or Lactobacillus and Bifidobacterium), type of vehicle used to deliver the probiotics (food vs. non-food (powder/capsule/tablet), and baseline BMI (< 30 kg/m2 vs. ≥ 30 kg/m2). The leave-one-out approach was used in the sensitivity analysis. Funnel plots and Egger’s test were used to appraise the possible publication bias in this study.

Results

Study characteristics

The database search yielded 4,048 records, and one additional record (a conference paper [23]) was obtained from the manual search of the references of the included RCTs. A total of 1,125 records were then excluded due to duplication, leaving 2,924 articles for screening. After the screening based on the titles and abstracts, 2,821 articles were further excluded (e.g. reviews, protocols, animal studies, etc.). The full texts of the remaining 103 potentially relevant studies were assessed according to the inclusion and exclusion criteria. Finally, thirty RCTs were included in this systematic review and meta-analysis (Fig. 1).
For the included 30 RCTs, all of them reported FBG, 17 RCTs reported HOMA-IR, 17 RCTs reported insulin, and 23 RCTs reported HbA1C (Fig. 1). Table 1 shows the basic information for the included 30 studies. Nine studies were conducted in Asian patients (three in China [15, 16, 24] and one each in India [25], Indonesia [26], Thailand [27], Japan [28], Malaysia [29], and Korea [30]), 19 studies were conducted in Caucasian patients (12 in Iran [11, 12, 3140] and one each in Ukraine [41], Turkey [23], Sweden [42], Saudi Arabia40 [43], Egypt [44], Denmark [45], and Australia [46]) and two studies were conducted in other races (two in Brazil [47, 48]). In the 30 RCTs, there were a total of 1,827 subjects, with 922 in the probiotic group and 905 in the control group. The dose of probiotics used in the 30 studies ranged from 2 × 107 to 1 × 1012 CFU/day, the duration of the probiotic interventions ranged from 4 to 36 weeks, and the baseline BMI ranged from 23.1 to 35.9 kg/m2. The probiotics were consumed as food (n = 13) or non-food (powder/capsule/tablet; n = 15) forms, and the probiotic genera were mainly Lactobacillus (n = 11), Bifidobacterium (n = 2), and Lactobacillus and Bifidobacterium (n = 14; Table 1).
Table 1
Characteristics of the included studies (n = 30)
First author, year
Country
Sample size
Age, years (mean ± SD)
BMI, kg/m2 (mean ± SD)
Study design
Type of vehicles for probiotics
Probiotics (genus and daily dose)
Duration
(weeks)
P
C
P
C
P
C
Toejing, 2021 [27]
Thailand
18
18
63.5 ± 5.9
61.8 ± 7.7
23.22 ± 2.72
23.05 ± 2.60
DB, PC
Powder
L. paracasei HII01 (5 × 1010 CFU/day)
12
Zhang, 2020 [15]
China
102
103
52.6 ± 10.5
53.6 ± 11.3
25.60 ± 2.96
26.20 ± 3.43
DB, PC
Powder
Probiotics mixture of (5 × 1010 CFU/day, Bifidobacterium longum BL88-Onlly, Bifido -bacterium breve BB8, Lactococcus gasseri LG23, Lactobacillus rhamnosus LR22, Lactobacillus salivarius LS86, Lactobacillus crispatus LCR15, Lactobacillus plantarum LP-Onlly, Lactobacillus fermentum LF33, Lactobacillus casei LC18)
13
Palacios, 2020 [46]
Australia
30
30
61.4 ± 8.9
56.1 ± 12.3
35.50 ± 6.20
36.30 ± 7.50
DB, PC
Capsule
Probiotics mixture of Lactobacillus plantarum Lp-115 1 (2 × 1010 CFU/day), Lacto -bacillus bulgaricus Lb-64 (6 × 109 CFU/day), Lactobacillus gasseri Lg-36 (3.6 × 1010 CFU/day), Bifidobacterium breve Bb-03 (1.5 × 1010 CFU/day), Bifidobacterium animalis sbsp. lactis Bi-07 (1.6 × 1010 CFU/day), Bifidobacterium bifidum Bb-06 (1.4 × 1010 CFU/day), Streptococcus thermophilus St-21 (9 × 108 CFU/day), Saccharomyces boulardii DBVPG 6763 (9 × 107 CFU /day)
12
Lsmail, 2020 [44]
Egypt
50
50
48.3 ± 12.9
46.4 ± 13.2
31.10 ± 5.30
30.20 ± 6.00
PC
Yogurt
Bifidobacterium animalis dn-173 010 (NR)
16
Tipici, 2020 [23]
Turkey
17
17
NR
NR
35.51 ± 7.33
33.65 ± 6.17
PC
NR
Lactobacillus GG (1 × 1010 CFU/day)
8
Jiang, 2020 [16]
China
42
34
56.0 ± 8.5
56.1 ± 8.2
27.51 ± 3.22
26.44 ± 2.78
DB, PC
Capsule
Probiotics mixture of (Bifidobacterium bifidum (1.2 × 109 CFU/day), Lactobacillus acidophilus (4.2 × 109 CFU/day), Streptococcus thermophilus (4.3 × 109 CFU/day))
12
Sabico, 2019 [43]
Saudi Arabia
31
30
48.0 ± 8.3
46.6 ± 5.9
30.10 ± 5.00
29.40 ± 5.20
DB, PC
Powder
Probiotics mixture of (4 × 109 CFU/day, Bifidobacterium bifidum W23, Bifidobacterium lactis W52, Lactobacillus acidophilus W37, Lactobacillus brevis W63, Lactobacillus casei W56, Lactobacillus salivarius W24, Lactococcus lactis W19 and Lactobacillus lactis W58)
24
Razmpoosh, 2019 [12]
Iran
30
30
58.6 ± 6.5
61.3 ± 5.2
27.70 ± 4.20
27.20 ± 4.20
DB, PC
Capsule
Probiotics mixture of (Lactobacillus acidophilus (4 × 109 CFU/day) Lactobacillus casei (1.4 × 1010 CFU/day), Lactobacillus rhamnosus (3 × 109 CFU/day), Lactobacillus bulgaricus (4 × 108 CFU/day), Bifidobacterium breve (6 × 1010 CFU/day), Bifidobacterium longum (1.4 × 1010 CFU/day), Streptococcus thermophilus (3 × 109 CFU/day))
6
Khalili, 2019 [33]
Iran
20
20
44.0 ± 8.1
45.0 ± 5.4
29.50 ± 3.34
31.94 ± 5.76
DB, PC
Capsule
cfu L. casei (1 × 108 CFU/day)
8
Madempudi, 2019 [25]
India
40
39
54.1
50.6
NR
NR
DB, PC
Capsule
Probiotics mixture of (6 × 1010 CFU/day, L. salivarius UBLS22, L. casei UBLC42, L. plantarum UBLP40, L. acidophilus UBLA34, B. breve UBBr01, and B. coagulans Unique IS2)
12
Raygan, 2018 [39]
Iran
30
30
60.7 ± 9.4
61.8 ± 9.8
30.30 ± 5.20
29.30 ± 4.10
DB, PC
Capsule
Probiotics mixture of (Bifidobacterium bifidum (2 × 109 CFU/day), Lactobacillus casei (2 × 109 CFU/day), Lactobacillus acidophilus (2 × 109 CFU/day))
12
Hsieh, 2018 [24]
China
22
22
52.3 ± 10.2
55.8 ± 8.6
28.04 ± 4.29
27.53 ± 3.15
DB, PC
Capsule
Lactobacillus reuteri ADR-1 (4 × 109 CFU/day)
36
Kobyliak, 2018 [41]
Ukraine
31
22
52.2 ± 1.7
57.18 ± 2.06
34.70 ± 1.29
35.65 ± 1.57
DB, PC
NR
Probiotics mixture of (Lactococcus (6 × 1011 CFU/day), Bifidobacterium (1 × 1011 CFU/day), Propionibacterium (3 × 1011 CFU/day), Acetobacter (1 × 107 CFU/day))
8
Sato, 2017 [28]
Japan
34
34
64.0 ± 9.2
65.0 ± 8.3
24.20 ± 2.60
24.60 ± 2.60
DB, PC
Fermented milk
Lactobacillus casei strain Shirota (4 × 1010 CFU/day)
16
Mobini, 2017 [42]
Sweden
14
15
64.0 ± 6.0
65.0 ± 5.0
32.30 ± 3.40
30.70 ± 4.00
DB, PC
Tablet
Lactobacillus reuteri DSM 17938 (1 × 1010 CFU/day)
12
Firouzi, 2017 [29]
Malaysia
48
53
52.9 ± 9.2
54.2 ± 8.3
29.20 ± 5.60
29.30 ± 5.30
DB, PC
Powder
Probiotics mixture of (6 × 1010 CFU/day, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus lactis, Bifidobacterium bifidum, Bifidobacterium longum, Bifidobacterium infantis)
12
Feizollahzadeh, 2017 [32]
Iran
20
20
56.9 ± 8.1
53.6 ± 7.2
26.68 ± 3.18
26.58 ± 3.24
DB, PC
Soy milk
Lactobacillus planetarum A7 (2 × 107 CFU/day)
8
Tonucci, 2017 [47]
Brazil
23
22
51.8 ± 6.6
51.0 ± 7.2
27.49 ± 3.97
27.94 ± 4.15
DB, PC
Fermented milk
Probiotics mixture of (Lactobacillus acidophilus La-5 (1 × 109 CFU/day), Bifidobacterium animalis subsp. lactis BB-12 (1 × 109 CFU/day))
6
Bayat, 2016 [31]
Iran
20
20
54.1 ± 9.5
47.0 ± 9.3
28.77 ± 4.59
29.75 ± 4.66
PC
Yogurt
NR
8
Ostadrahimi, 2015 [48]
Brazil
30
30
NR
NR
28.89 ± 4.77
27.47 ± 3.55
DB, PC
Fermented milk
Bifidobacterium animalis HN019 2.7 (2 × 1010 CFU/day)
8
Hove, 2015 [45]
Denmark
23
18
58.5 ± 7.7
60.6 ± 5.2
29.20 ± 3.80
27.70 ± 3.30
DB, PC
Fermented milk
Lactobacillus helveticus Cardi04 (NR)
12
Tajadadi-Ebrahimi, 2014 [36]
Iran
27
27
52.0 ± 7.2
53.4 ± 7.5
29.80 ± 5.70
30.50 ± 4.10
DB, PC
Bread
Lactobacillus Sporogenes (3 × 108 CFU/day)
8
Shakeri, 2014 [35]
Iran
26
26
52.3 ± 8.2
53.1 ± 7.5
29.50 ± 5.70
30.60 ± 4.10
DB, PC
Bread
L. Sporogenes (1.2 × 1010 CFU/day)
8
Mohamadshahi, 2014 [34]
Iran
22
22
53.0 ± 5.9
49.0 ± 7.1
28.36 ± 4.14
29.22 ± 3.20
DB, PC
Yogurt
Probiotics mixture of (Lactobacillus acidophilus (1.11 × 109 CFU/day), Bifidobacterium lactic (1.11 × 109 CFU / day))
8
Jung, 2014 [30]
Korea
21
20
63.3 ± 9.2
60.2 ± 8.5
25.90 ± 4.12
25.60 ± 3.13
DB, PC
Milk
probiotics mixture of (Lactobacillus acidophilus (3 × 1010 CFU/day), Lactobacillus casei (1.8 × 1010 CFU/day), Bifidobacterium lactis (9.6 × 1010 CFU/day), Streptococcus thermophilus (NR))
8
Judiono, 2014 [26]
Indonesian
36
36
NR
NR
NR
NR
PC
Milk
Probiotics mixture of (lactic acid bacterias (2 × 109 CFU/day), and other 34 beneficial heal -thy probiotic bacterias (NR))
4
Mazloom, 2013 [38]
Iran
16
18
55.4 ± 8.0
51.8 ± 10.2
27.97 ± 3.81
27.24 ± 2.73
SB, PC
Capsule
Probiotics mixture of (Lactobacillus acidophilus (NR), Lactobacillus bulgaricus (NR), Lactobacillus bififi dum (NR), Lactobacillus casei (NR))
6
Asemi, 2013 [11]
Iran
27
27
50.5 ± 9.8
52.6 ± 7.1
31.61 ± 6.36
30.17 ± 4.23
DB, PC
Capsule
probiotics mixture of (L. acidophilus (2 × 109 CFU/day), L. casei (7 × 109 CFU/day), L. rha -mnosus (1.5 × 109 CFU/day), L. bulgaricus (2 × 108 CFU/day), Bifidobacterium breve (2 × 1010 CFU/day), B. longum (7 × 109 CFU/day), Streptococcus thermophilus (1.5 × 109 CFU/day))
8
Hosseinzadeh, 2013 [40]
Iran
42
42
46.8 ± 6.2
45.7 ± 6.1
30.00 ± 4.40
29.90 ± 4.70
DB, PC
Tablet
NR
12
Ejtahed, 2012 [37]
Iran
30
30
50.9 ± 7.7
51.0 ± 7.3
28.95 ± 3.65
29.14 ± 4.30
DB, PC
Yogurt
Probiotics mixture of (Lactobacillus acidophilus La5 (2.2 × 109 CFU), Bifidobacterium lactis Bb12 (1.8 × 109 CFU), Lactobacillus bulgaricus (NR), Streptococcus thermophilus (NR))
6
P, probiotic group; C, control group; DB, double-blinded; PC, placebo-controlled; SB, single blinded; NR, not reported; CFU, colony-forming units

Risk of bias assessment of the included RCTs

The Cochrane risk-of-bias assessment tool was used to assess the bias of the 30 included studies. Approximately half of the studies (53%) were randomised, but 14 studies did not clearly report the randomisation process. The methods of allocation concealment were described in 43% of the included RCTs, and the majority of the studies (87%) described the blinding method. Approximately 40% of the studies provided information about the blinding outcome assessment. Most of the included studies had a low risk of attrition bias (73%), a low risk of reporting bias (93%), and a low risk of other types of bias (70%). Overall, four of the studies were classified as high quality (all terms were assessed as low risk), 19 studies were classified as moderate quality (no term was assessed as a high risk and one or more terms were assessed as unclear risks), and seven studies were classified as low quality (one or more terms were assessed as a high risk). The general and individual risks of bias are shown in Additional file 3: Fig. S1.

Effects of probiotic supplementation on glycaemic control

Effects on FBG

Thirty studies including a total of 1,827 T2DM patients were used to evaluate the effects of probiotic supplementation on FBG level. The pooled effects of probiotic supplementation indicated a significant decrease in FBG level in the probiotic group (SMD = − 0.331, 95% CI  − 0.424 to − 0.238, Peffect < 0.001), and the heterogeneity was low (I2 = 29%, Pheterogeneity = 0.070; Fig. 2a). Leave-one-out sensitivity analysis confirmed that the pooled effects of probiotic supplementation on FBG level were stable and reliable (Additional file 4: Fig. S2a).
Subgroup analyses for FBG were performed according to race, probiotic intervention dose, probiotics genus, type of vehicle used to deliver the probiotics, and baseline BMI. As shown in Table 2, the significant subgroup differences (Psubgroup < 0.050) were observed for races (Asian vs. Caucasian), genus of probiotics (Lactobacillus vs. Bifidobacterium vs. Lactobacillus and Bifidobacterium), and baseline BMI (< 30 kg/m2 vs. ≥ 30 kg/m2). A stronger beneficial effect of the probiotic intervention was observed on FBG level in the Caucasian subgroup (SMD = − 0.448, 95% CI  − 0.575 to − 0.322, Peffect < 0.001, Psubgroup = 0.020), in the Bifidobacterium subgroup (SMD = − 0.626, 95% CI  − 1.221 to − 0.030, Peffect = 0.039, Psubgroup = 0.040), and in the high-baseline-BMI (≥ 30 kg/m2) subgroup (SMD = -0.490, 95% CI  − 0.644 to − 0.336, Peffect < 0.001, Psubgroup = 0.007). No differences were observed between the subgroups of probiotic dose, intervention duration, or type of vehicle used to deliver the probiotics (Table 2, Psubgroup > 0.050).
Table 2
Subgroup analysis for the effects of probiotics on FBG
Subgroup
No. of trials
No. of participants
I2 (%)
Pheterogeneity
Pooled SMD [95% CI]
Peffect
Psubgroup
Race
       
 Asian
9
722
18.5
0.279
− 0.217 [− 0.364, − 0.070]
0.004
0.020
 Caucasian
19
1000
24.8
0.157
− 0.448 [− 0.575, − 0.322]
 < 0.001
Dose of probiotics
       
  ≤ 1 × 1010 CFU/day
12
643
38.8
0.082
− 0.335 [− 0.500, − 0.170]
0.003
0.412
  > 1 × 1010 CFU/day
13
885
0.0
0.203
− 0.248 [− 0.376, − 0.119]
 < 0.001
Duration of intervention
       
  ≤ 8 weeks
16
783
0.0
0.541
− 0.300 [− 0.441, − 0.158]
 < 0.001
0.506
  > 8 weeks
14
1044
52.9
0.010
− 0.401 [− 0.588, − 0.215]
 < 0.001
Genus of probiotics
       
 Lactobacillus
11
472
0.0
0.669
− 0.194 [− 0.376, − 0.012]
0.037
0.040
 Bifidobacterium
2
160
69.9
0.068
− 0.626 [− 1.221, − 0.030]
0.039
 Lactobacillus and Bifidobacterium
15
1071
32.1
0.111
− 0.346 [− 0.498, − 0.195]
 < 0.001
Type of vehicle used to deliver the probiotics
       
 Powder/capsule/tablet
15
1023
36.4
0.078
− 0.357 [− 0.481, − 0.232]
 < 0.001
0.809
 Food
13
717
35.1
0.102
− 0.333 [− 0.481, − 0.184]
 < 0.001
Baseline BMI
       
  < 30 kg/m2
16
995
2.8
0.420
− 0.218 [− 0.343, − 0.092]
0.001
0.007
  ≥ 30 kg/m2
12
681
40.6
0.070
− 0.490 [− 0.644, − 0.336]
 < 0.001
FBG, fasting blood glucose; CFU, colony-forming units; BMI,body mass index

Effects on insulin

Eight hundred and eighty-six patients in 17 RCTs were included in the meta-analysis of the effects of probiotic intake on insulin level. Probiotic supplementation in T2DM patients led to a significant reduction in insulin level (SMD = − 0.185, 95% CI  − 0.313 to − 0.056, Peffect = 0.004) without heterogeneity (Fig. 2b, I2 = 0%, Pheterogeneity = 0.500). Sensitivity analysis also supported the robustness of the results for insulin level (Additional file 4: Fig. S2b).
As shown in Table 3, the magnitude of the reduction was significantly greater in the subgroup of patients taking food-type probiotics (SMD = − 0.386, 95% CI  − 0.592 to − 0.180, Peffect < 0.001, Psubgroup = 0.014) than in the subgroup taking non-food (powder/capsule/tablet) types. In addition, no differences were observed between the subgroups of races, probiotic dose, intervention duration, probiotic genus, or baseline BMI (Table 3, Psubgroup > 0.050).
Table 3
Subgroup analysis for the effects of probiotics on insulin
Subgroup
No. of trials
No. of participants
I2 (%)
Pheterogeneity
Pooled SMD [95% CI]
Peffect
Psubgroup
Race
       
 Asian
5
313
0.0
0.777
− 0.145 [− 0.367, 0.078]
0.202
0.764
 Caucasian
11
513
23.3
0.222
− 0.187 [− 0.352, − 0.022]
0.027
Dose of probiotics
       
  ≤ 1 × 1010 CFU/day
9
516
27.2
0.202
− 0.169 [− 0.343, 0.005]
0.056
0.851
  > 1 × 1010 CFU/day
6
323
0.0
0.673
− 0.143 [− 0.362, 0.076]
0.202
Duration of intervention
       
  ≤ 8 weeks
10
478
0.0
0.693
− 0.291 [− 0.463, − 0.120]
0.001
0.071
  > 8 weeks
7
408
1.1
0.416
− 0.052 [− 0.247, 0.143]
0.600
Genus of probiotics
       
 Lactobacillus
6
356
21.1
0.275
− 0.300 [− 0.510, − 0.090]
0.005
0.183
 Lactobacillus and Bifidobacterium
10
584
0.0
0.661
− 0.119 [− 0.282, 0.044]
0.152
Type of vehicle used to deliver the probiotics
       
 Powder/capsule/tablet
10
510
0.0
0.630
− 0.049 [− 0.223, 0.125]
0.581
0.014
 Food
6
316
0.0
0.744
− 0.386 [− 0.592, − 0.180]
 < 0.001
Baseline BMI
       
  < 30 kg/m2
8
413
0.0
0.732
− 0.261 [− 0.455, − 0.066]
0.009
0.279
  ≥ 30 kg/m2
8
448
31.2
0.179
− 0.112 [− 0.299, 0.075]
0.239
CFU, colony-forming units; BMI,body mass index

Effects on HbA1c

The effects of probiotic interventions on HbA1c level were evaluated in 23 RCTs including 1,466 T2DM patients. A significant decrease was observed in the HbA1c level in the probiotic group (Fig. 2c, SMD = − 0.421, 95% CI  − 0.583 to − 0.258, Peffect < 0.001) with moderate heterogeneity (I2 = 56%, Pheterogeneity < 0.001). Sensitivity analysis showed that the results for HbA1 level were stable and reliable (Additional file 4: Fig. S2c).
The subgroup analysis was performed for HbA1c according to races (Asian vs. Caucasian), genera of probiotics (Lactobacillus vs. Bifidobacterium vs. Lactobacillus and Bifidobacterium), types of vehicle used to deliver the probiotics (food vs. non-food (powder/capsule/tablet)), and baseline BMI (< 30 kg/m2 vs. ≥ 30 kg/m2). As shown in Table 4, a significantly greater reduction was observed in the HbA1c level in the subgroups of Caucasians (SMD = − 0.465, 95% CI  − 0.672 to − 0.257, Peffect < 0.001, Psubgroup = 0.032), Bifidobacterium probiotics (SMD = − 0.913, 95% CI  − 1.387 to − 0.438, Peffect < 0.001, Psubgroup = 0.001), food-type probiotics (SMD = − 0.524, 95% CI  − 0.800 to − 0.249, Peffect < 0.001, Psubgroup = 0.047), and baseline BMI ≥ 30 kg/m2 (SMD = − 0.485, 95% CI  − 0.783 to − 0.188, Peffect = 0.001, Psubgroup = 0.018). No differences were observed between the subgroups of probiotic dose or intervention duration (Psubgroup > 0.050).
Table 4
Subgroup analysis for the effects of probiotics on HbA1c
Subgroup
No. of trials
No. of participants
I2 (%)
Pheterogeneity
Pooled SMD [95% CI]
Peffect
Psubgroup
Race
       
 Asian
9
722
70.2
0.001
− 0.362 [− 0.647, − 0.077]
0.013
0.032
 Caucasian
12
639
39.6
0.077
− 0.465 [− 0.672, − 0.257]
 < 0.001
Dose of probiotic
       
  ≤ 1 × 1010 CFU/day
8
368
5.0
0.392
− 0.337 [− 0.573, − 0.102]
 < 0.001
0.076
 > 1 × 1010 CFU/day
11
833
62.8
0.003
− 0.484 [− 0.699, − 0.270]
0.005
Duration of intervention
       
  ≤ 8 weeks
11
543
0.0
0.586
− 0.509 [− 0.681, − 0.337]
 < 0.001
0.077
  > 8 weeks
12
923
72.4
 < 0.001
− 0.359 [− 0.619, − 0.098]
0.007
Genus of probiotics
       
 Lactobacillus
7
292
54.1
0.042
− 0.250 [− 0.599, 0.098]
0.159
0.001
 Bifidobacterium
2
160
50.7
0.154
− 0.913 [− 1.387, − 0.438]
 < 0.001
 Lactobacillus and Bifidobacterium
12
890
51.0
0.021
− 0.407 [− 0.605, − 0.209]
 < 0.001
 
Type of vehicle used to deliver the probiotics
       
 Powder/capsule/tablet
11
808
55.9
0.012
− 0.384 [− 0.606, − 0.162]
0.001
0.047
 Food
10
571
61.5
0.005
− 0.524 [− 0.800, − 0.249]
 < 0.001
Baseline BMI
       
  < 30 kg/m2
13
861
51.1
0.017
− 0.338 [− 0.541, − 0.134]
0.001
0.018
  ≥ 30 kg/m2
8
454
57.6
0.021
− 0.485 [− 0.783, − 0.188]
0.001
HbA1c, haemoglobin A1c; CFU, colony-forming units; BMI,body mass index

Effects on the HOMA-IR score

The results of the meta-analysis of 17 RCTs (n = 1,116) suggested significant effects of probiotic interventions on reducing the HOMA-IR scores in T2DM patients (SMD = − 0.224, 95% CI  − 0.342 to − 0.105, Peffect < 0.001). The heterogeneity (I2 = 41%, Pheterogeneity = 0.040) of these RCTs was moderate (Fig. 2b). Sensitivity analysis showed that the pooled effects of probiotic supplementation on HOMA-IR scores did not significantly change, suggesting that the meta-analysis results were stable and reliable (Additional file 4: Fig. S2b).
No statistically significant differences were observed in the HOMA-IR score between subgroups (Table 5, Psubgroup > 0.050). However, an effective reduction in the HOMA-IR score was observed in the subgroups of Caucasians (SMD = − 0.308, 95% CI  − 0.471 to − 0.146, Peffect < 0.001, Psubgroup = 0.173), high baseline BMI (≥ 30 kg/m2; SMD = − 0.320, 95% CI  − 0.615 to − 0.026, Peffect = 0.033, Psubgroup = 0.144), and Bifidobacterium probiotics (SMD = − 0.248, 95% CI  − 0.387 to − 0.109, Peffect = 0.004, Psubgroup = 0.345).
Table 5
Subgroup analysis for the effects of probiotics on HOMA-IR
Subgroup
No. of trials
No. of participants
I2 (%)
Pheterogeneity
Pooled SMD [95% CI]
Peffect
Psubgroup
Race
       
 Asian
5
470
39.0
0.161
− 0.139 [− 0.321, 0.043]
0.134
0.173
 Caucasian
11
601
47.3
0.040
− 0.308 [− 0.471, − 0.146]
 < 0.001
Dose of probiotics
       
  ≤ 1 × 1010 CFU/day
6
304
69.2
0.006
− 0.179 [− 0.593, 0.234]
0.396
0.969
  > 1 × 1010 CFU/day
8
653
33.3
0.163
− 0.241 [− 0.438, − 0.043]
0.017
Duration of intervention
       
  ≤ 8 weeks
8
381
32.7
0.167
− 0.163 [− 0.412, 0.085]
0.198
0.496
  > 8 weeks
9
735
52.4
0.032
− 0.312 [− 0.534, − 0.091]
0.006
Genus of probiotics
       
 Lactobacillus
5
213
43.8
0.130
− 0.101 [− 0.373, 0.170]
0.606
0.345
 Lactobacillus and Bifidobacterium
11
819
48.4
0.036
− 0.248 [− 0.387, − 0.109]
0.004
Type of vehicle used to deliver the probiotics
       
 Powder/capsule/tablet
12
882
57.7
0.007
− 0.239 [− 0.453, − 0.026]
0.028
0.912
 Food
4
181
0.0
0.829
− 0.230 [− 0.523, 0.063]
0.124
Baseline BMI
       
  < 30 kg/m2
8
571
11.9
0.338
− 0.165 [− 0.347, 0.017]
0.075
0.144
  ≥ 30 kg/m2
8
466
60.0
0.015
− 0.320 [− 0.615, − 0.026]
0.033
HOMA-IR, homeostasis model of assessment of insulin resistance, CFU, colony-forming units; BMI,body mass index

Publication bias analysis

Potential publication bias was assessed using funnel plots and Egger’s test. A visual inspection of the funnel plots revealed no publication bias for FBG, insulin, or HbA1c levels or the HOMA-IR score (Additional file 5: Fig. S3). Egger’s test results showed no publication bias for FBG (P = 0.349), insulin (P = 0.260) or HbA1c (P = 0.108) levels or the HOMA-IR score (P = 0.391).

Discussion

This systematic review and meta-analysis summarised data from 30 RCTs, including a total of 1,827 individuals, to evaluate the effects of probiotic supplementation on glycaemic control in T2DM patients. The results revealed that probiotic supplementation significantly decreased FBG, insulin, and HbA1c levels and HOMA-IR scores in T2DM patients. Further subgroup analyses showed that the effect was larger in the subgroups of Caucasians, high baseline BMI (≥ 30.0 kg/m2), Bifidobacterium probiotics, and food-type probiotics.
This study supported the notion that probiotics improve glycaemic control in T2DM patients. This is inconsistent with the results reported by the systematic review and meta-analysis of 12 RCTs in 2016 [13]. They found no significant differences in the HbA1c level and HOMA-IR score between the probiotic and control groups of T2DM patients. For their study, the meta-analysis of HbA1c and HOMA-IR were conducted with limited number of RCTs (n = 6), and five of them had the participants’ baseline BMI less than 30 kg/m2. In this study, the subgroup analysis found that the effect was larger in individuals with higher baseline BMI (≥ 30.0 kg/m2). These may partially explained the differences between the two studies. The gut microbiota is largely involved in the metabolic, nutritional, physiological, and immune functions of the host [4951]. A previous study showed that T2DM patients are characterised by a decrease in the abundance of certain butyrate-producing bacteria and the enrichment of other microbial functions conferring sulphate reduction and oxidative stress resistance [52]. Changes in the gut microbial composition may be a mechanism whereby probiotic supplementation improves glycaemic control. Probiotic supplementation may modulate and increase the abundance of intestinal flora that are beneficial to glycaemic control [53, 54]. Moreover, the gut microbiota may regulate glucagon-like peptide 1, which promotes the secretion of insulin from islet β cells, and reduces the secretion of glucagon from islet α cells, resulting in a reduction in gastric emptying time, gastrointestinal peristalsis, and loss of appetite [55, 56]. Previous studies have found that probiotics may stimulate the production of short-chain fatty acids, especially butyrate, which increase insulin sensitivity and thus improve glycaemic control [5759].
The subgroup analyses suggested that Bifidobacterium have greater effects than other probiotic genera. Probiotics that colonise the gut may change the host’s gut microbiota. According to a 5-year follow-up study, Bifidobacterium longum, a member of the core microbiota of the human gut, can stably colonise the gut [60]. Another study reported that oral supplementation with B. longum persists in the gut for 6 months in 30% of subjects [61]. Moreover, Xiao et al. (2020) found that Bifidobacterium appears to have a better ability to colonise the gut than Lactobacillus [62]. This may explain the finding that Bifidobacterium had a larger effect than other probiotic genera on glycaemic-control-related parameters (e.g. FBG and HbA1c levels) in T2DM patients, to some extent, in this study.
Food-type probiotics (e.g. yogurt and fermented milk) may have greater effects than other types of probiotics on glycaemic control in T2DM patients. Gastric acidity is thought to be one of the main obstacles to gut colonisation [63, 64]. Food-type probiotics (e.g. yogurt and fermented milk) may buffer the stomach acid, allowing the probiotics to better colonise the gut [65]. An in vitro study assessed the tolerance of probiotics in the human gastrointestinal tract by evaluating the effects of food addition on the viability of probiotics in simulated pH 2.0 gastric juices, revealing that adding soymilk or a liquid breakfast greatly enhanced the survival of the probiotics [66].
Compared to the baseline BMI < 30 kg/m2 subgroup, the stronger beneficial effects of a probiotic intervention were also observed on FBG and HbA1c levels in the baseline BMI ≥ 30.0 kg/m2 subgroup. This may be due to gut dysbiosis in obese individuals. In 2021, Liu et al. summarised the characteristics of the gut microbiota in obesity. Obese individuals were observed to have an increased Firmicutes/Bacteroidetes ratio at the phylum level and decreased abundances of the genera Lactobacillus and Bifidobacterium [67]. Probiotic supplementation may alleviate gut dysbiosis [68]. These findings indicate that obese individuals may be more sensitive to probiotic interventions. In addition, this may partly explain the observed racial differences, i.e. the effect was larger in Caucasians than in Asians. In this study, the average baseline BMI (30.3 kg/m2) was higher in Caucasians than in Asians (26.2 kg/m2).
In addition, no significant difference was observed between the longer-term intervention (> 8 weeks) and the shorter-term intervention (≤ 8 weeks) groups. In 2020, an RCT was conducted in 150 new-borns (38–40 weeks gestational age). In that study, the intervention group received probiotic supplementation containing 2 × 106 CFU/day of B. breve PB04 and L. rhamnosus KL53A. The stool samples from days 5, 6, and 30 were collected for an analysis of the gut microbiome. The results showed that L. rhamnosus and B. breve colonised rapidly, generally on days 5 and 6 [69]. This ability of the probiotics to rapidly colonise the gut may have resulted in the very small difference between the short and long intervention durations.
Furthermore, no significant differences were found between the higher-dose (> 1 × 1010 CFU/day) and lower-dose (≤ 1 × 1010 CFU/day) probiotic intervention groups. Several studies have reported similar results. Ibarra et al. (2018) performed a randomised double-blind, placebo-controlled trial to determine the effects of 4 weeks of supplementation with 1 × 109 or 1 × 1010 CFU of B. animalis subsp. lactis HN019 on adults diagnosed with functional constipation. The results showed no significant difference between the two groups with different doses of probiotics [70]. However, Whorwell et al. (2006) conducted a multi-centre clinical trial of 362 patients with irritable bowel syndrome (IBS) and found that 1 × 108 CFU of B. infantis 35,624 significantly alleviated the symptoms of IBS and that its effect was superior to that of the administration of 1 × 106 CFU/day and 1 × 1010 CFU/day of B. infantis 35624 [71]. In all of the included RCTs, the probiotic intervention doses were higher than 1 × 106 CFU/day, and only one RCT had a probiotic intervention dose lower than 1 × 108 CFU/day. Thus, these two doses were not used as the limits for subgroup analysis in this systematic review and meta-analysis. Further studies are warranted to determine the optimal dose of probiotics for glycaemic control in T2DM patients.
This study systematically and comprehensively evaluated the effects of probiotic supplementation on glycaemic control in T2DM patients. To the best of our knowledge, this is the first systematic review and meta-analysis study to investigate the differences in the effects of probiotic interventions on glycaemic control in T2DM patients according to participant characteristics (e.g. race, baseline BMI), and intervention characteristics, (e.g. probiotic doses, probiotic genus, treatment duration, and types of vehicles used to deliver the probiotics). However, this study also has some limitations. First, as 12 of the included studies (40%) were conducted in Iran, some racial and ethnic groups may be underrepresented. This may have resulted in a limited racial representation. Second, the number of RCTs in some subgroup analyses was low. For example, in the subgroup analysis of HbA1c level, the number of RCTs in the Bifidobacterium subgroup was only two. Third, the duration of most of the RCTs included in the analysis was from 4 to 24 weeks, and only one RCT was longer than 24 weeks (a 36-week intervention). Therefore, the long-term effects could not be explored in this study.

Conclusions

The findings of this study indicate that probiotic supplementation had favourable effects on glycaemic control in T2DM patients. Bifidobacterium and food-type probiotics had greater glucose-lowering effects than other probiotic genera and types of vehicle used to deliver the probiotics. Patients with a higher BMI may gain more glycaemic control benefits from a probiotic intervention. The administration of probiotics may be a promising adjuvant therapy for glycaemic control in T2DM patients.

Acknowledgements

None.

Declarations

Not applicable.
Not applicable.

Competing interests

All the authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Alvarez-Silva C, Kashani A, Hansen TH, et al. Trans-ethnic gut microbiota signatures of type 2 diabetes in Denmark and India. Genome Med. 2013;13(1):37.CrossRef Alvarez-Silva C, Kashani A, Hansen TH, et al. Trans-ethnic gut microbiota signatures of type 2 diabetes in Denmark and India. Genome Med. 2013;13(1):37.CrossRef
2.
Zurück zum Zitat Sun H, Saeedi P, Karuranga S, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183: 109119.PubMedCrossRef Sun H, Saeedi P, Karuranga S, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183: 109119.PubMedCrossRef
4.
Zurück zum Zitat Heald AH, Stedman M, Davies M, et al. Estimating life years lost to diabetes: outcomes from analysis of National Diabetes Audit and Office of National Statistics data. Cardiovasc Endocrinol Metab. 2020;9(4):183–5.PubMedPubMedCentralCrossRef Heald AH, Stedman M, Davies M, et al. Estimating life years lost to diabetes: outcomes from analysis of National Diabetes Audit and Office of National Statistics data. Cardiovasc Endocrinol Metab. 2020;9(4):183–5.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Ng SC, Xu Z, Mak JWY, et al. Microbiota engraftment after faecal microbiota transplantation in obese subjects with type 2 diabetes: a 24-week, double-blind, randomised controlled trial. Gut. 2022;71(4):716–23.PubMedCrossRef Ng SC, Xu Z, Mak JWY, et al. Microbiota engraftment after faecal microbiota transplantation in obese subjects with type 2 diabetes: a 24-week, double-blind, randomised controlled trial. Gut. 2022;71(4):716–23.PubMedCrossRef
6.
Zurück zum Zitat Umirah F, Neoh CF, Ramasamy K, Lim SM. Differential gut microbiota composition between type 2 diabetes mellitus patients and healthy controls: a systematic review. Diabetes Res Clin Pract. 2021;173: 108689.PubMedCrossRef Umirah F, Neoh CF, Ramasamy K, Lim SM. Differential gut microbiota composition between type 2 diabetes mellitus patients and healthy controls: a systematic review. Diabetes Res Clin Pract. 2021;173: 108689.PubMedCrossRef
7.
Zurück zum Zitat Mcfarland LV. A review of the evidence of health claims for biotherapeutic agents. Microb Ecol Health Dis. 2009;12:65–76. Mcfarland LV. A review of the evidence of health claims for biotherapeutic agents. Microb Ecol Health Dis. 2009;12:65–76.
8.
Zurück zum Zitat Salminen S. Human studies on probiotics: aspects of scientific documentation. Näringsforskning. 2001;45(1):8–12.CrossRef Salminen S. Human studies on probiotics: aspects of scientific documentation. Näringsforskning. 2001;45(1):8–12.CrossRef
9.
Zurück zum Zitat Yang K, Niu J, Zuo T, et al. Alterations in the gut virome in obesity and type 2 diabetes mellitus. Gastroenterology. 2021;161(4):1257-1269.e13.PubMedCrossRef Yang K, Niu J, Zuo T, et al. Alterations in the gut virome in obesity and type 2 diabetes mellitus. Gastroenterology. 2021;161(4):1257-1269.e13.PubMedCrossRef
10.
Zurück zum Zitat Barrea L, Vetrani C, Verde L, et al. Comprehensive approach to medical nutrition therapy in patients with type 2 diabetes mellitus: from diet to bioactive compounds. Antioxidants (Basel). 2023;12(4):904.PubMedCrossRef Barrea L, Vetrani C, Verde L, et al. Comprehensive approach to medical nutrition therapy in patients with type 2 diabetes mellitus: from diet to bioactive compounds. Antioxidants (Basel). 2023;12(4):904.PubMedCrossRef
11.
Zurück zum Zitat Asemi Z, Zare Z, Shakeri H, Sabihi SS, Esmaillzadeh A. Effect of multispecies probiotic supplements on metabolic profiles, hs-CRP, and oxidative stress in patients with type 2 diabetes. Ann Nutr Metab. 2013;63(1–2):1–9.PubMedCrossRef Asemi Z, Zare Z, Shakeri H, Sabihi SS, Esmaillzadeh A. Effect of multispecies probiotic supplements on metabolic profiles, hs-CRP, and oxidative stress in patients with type 2 diabetes. Ann Nutr Metab. 2013;63(1–2):1–9.PubMedCrossRef
12.
Zurück zum Zitat Razmpoosh E, Javadi A, Ejtahed HS, Mirmiran P, Javadi M, Yousefinejad A. The effect of probiotic supplementation on glycemic control and lipid profile in patients with type 2 diabetes: a randomized placebo controlled trial. Diabetes Metab Syndr. 2019;13(1):175–82.PubMedCrossRef Razmpoosh E, Javadi A, Ejtahed HS, Mirmiran P, Javadi M, Yousefinejad A. The effect of probiotic supplementation on glycemic control and lipid profile in patients with type 2 diabetes: a randomized placebo controlled trial. Diabetes Metab Syndr. 2019;13(1):175–82.PubMedCrossRef
13.
Zurück zum Zitat Li C, Li X, Han H, et al. Effect of probiotics on metabolic profiles in type 2 diabetes mellitus: a meta-analysis of randomized, controlled trials. Medicine. 2017;95(26):e4088.CrossRef Li C, Li X, Han H, et al. Effect of probiotics on metabolic profiles in type 2 diabetes mellitus: a meta-analysis of randomized, controlled trials. Medicine. 2017;95(26):e4088.CrossRef
14.
15.
Zurück zum Zitat Zhang Y, Gu Y, Ren H, et al. Gut microbiome-related effects of berberine and probiotics on type 2 diabetes (the PREMOTE study). Nat Commun. 2020;11(1):5015.PubMedPubMedCentralCrossRef Zhang Y, Gu Y, Ren H, et al. Gut microbiome-related effects of berberine and probiotics on type 2 diabetes (the PREMOTE study). Nat Commun. 2020;11(1):5015.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Jiang H, Zhang Y, Xu D, Wang Q. Probiotics ameliorates glycemic control of patients with diabetic nephropathy: a randomized clinical study. J Clin Lab Anal. 2021;35(4): e23650.PubMedPubMedCentralCrossRef Jiang H, Zhang Y, Xu D, Wang Q. Probiotics ameliorates glycemic control of patients with diabetic nephropathy: a randomized clinical study. J Clin Lab Anal. 2021;35(4): e23650.PubMedPubMedCentralCrossRef
17.
18.
Zurück zum Zitat Higgins JPTTJ, Chandler J, Cumpston M, Li T, Page MJ, Welch VA. Cochrane handbook for systematic reviews of interventions. Chapter 16.1. Chichester: Wiley; 2019. p. 470–81.CrossRef Higgins JPTTJ, Chandler J, Cumpston M, Li T, Page MJ, Welch VA. Cochrane handbook for systematic reviews of interventions. Chapter 16.1. Chichester: Wiley; 2019. p. 470–81.CrossRef
19.
Zurück zum Zitat Follmann D, Elliott P, Suh I, Cutler J. Variance imputation for overviews of clinical trials with continuous response. J Clin Epidemiol. 1992;45(7):769–73.PubMedCrossRef Follmann D, Elliott P, Suh I, Cutler J. Variance imputation for overviews of clinical trials with continuous response. J Clin Epidemiol. 1992;45(7):769–73.PubMedCrossRef
20.
Zurück zum Zitat Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the sample size, median, mid-range, and/or mid-quartile range. Stat Methods Med Res. 2018;27(6):1785–805.PubMedCrossRef Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the sample size, median, mid-range, and/or mid-quartile range. Stat Methods Med Res. 2018;27(6):1785–805.PubMedCrossRef
21.
Zurück zum Zitat Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol. 2014;14:135.PubMedPubMedCentralCrossRef Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol. 2014;14:135.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Cohen J. Statistical power analysis for the behavioral sciences, chapter 10. Mahwah: Lawrence Erlbaum Associates; 1998. p. 467–81. Cohen J. Statistical power analysis for the behavioral sciences, chapter 10. Mahwah: Lawrence Erlbaum Associates; 1998. p. 467–81.
23.
Zurück zum Zitat Tipici BE, Coskunpinar E, Altunkanat D, et al. Effects of Lactobacillus GG supplementation in type 2 diabetes: Are mucin genes expressions important? Diabetologia. 2020;1:63. Tipici BE, Coskunpinar E, Altunkanat D, et al. Effects of Lactobacillus GG supplementation in type 2 diabetes: Are mucin genes expressions important? Diabetologia. 2020;1:63.
24.
Zurück zum Zitat Hsieh MC, Tsai WH, Jheng YP, et al. The beneficial effects of Lactobacillus reuteri ADR-1 or ADR-3 consumption on type 2 diabetes mellitus: a randomized, double-blinded, placebo-controlled trial. Sci Rep. 2018;8(1):16791.PubMedPubMedCentralCrossRef Hsieh MC, Tsai WH, Jheng YP, et al. The beneficial effects of Lactobacillus reuteri ADR-1 or ADR-3 consumption on type 2 diabetes mellitus: a randomized, double-blinded, placebo-controlled trial. Sci Rep. 2018;8(1):16791.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Madempudi RS, Ahire JJ, Neelamraju J, Tripathi A, Nanal S. Efficacy of UB0316, a multi-strain probiotic formulation in patients with type 2 diabetes mellitus: a double blind, randomized, placebo controlled study. PLoS ONE. 2019;14(11): e0225168.PubMedPubMedCentralCrossRef Madempudi RS, Ahire JJ, Neelamraju J, Tripathi A, Nanal S. Efficacy of UB0316, a multi-strain probiotic formulation in patients with type 2 diabetes mellitus: a double blind, randomized, placebo controlled study. PLoS ONE. 2019;14(11): e0225168.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Judiono J, Hadisaputro S, Indranila KS, et al. Effects of clear kefir on biomolecular aspects of glycemic status of type 2 diabetes mellitus (T2DM) patients in Bandung, West Java. Funct Foods Health Dis. 2014;4(8):340–8.CrossRef Judiono J, Hadisaputro S, Indranila KS, et al. Effects of clear kefir on biomolecular aspects of glycemic status of type 2 diabetes mellitus (T2DM) patients in Bandung, West Java. Funct Foods Health Dis. 2014;4(8):340–8.CrossRef
27.
Zurück zum Zitat Toejing P, Khampithum N, Sirilun S, Chaiyasut C, Lailerd N. Influence of Lactobacillus paracasei HII01 supplementation on glycemia and inflammatory biomarkers in type 2 diabetes: a randomized clinical trial. Foods (Basel, Switzerland). 2021;10(7):1455.PubMedPubMedCentral Toejing P, Khampithum N, Sirilun S, Chaiyasut C, Lailerd N. Influence of Lactobacillus paracasei HII01 supplementation on glycemia and inflammatory biomarkers in type 2 diabetes: a randomized clinical trial. Foods (Basel, Switzerland). 2021;10(7):1455.PubMedPubMedCentral
28.
Zurück zum Zitat Sato J, Kanazawa A, Azuma K, et al. Probiotic reduces bacterial translocation in type 2 diabetes mellitus: a randomised controlled study. Sci Rep. 2017;7(1):12115.PubMedPubMedCentralCrossRef Sato J, Kanazawa A, Azuma K, et al. Probiotic reduces bacterial translocation in type 2 diabetes mellitus: a randomised controlled study. Sci Rep. 2017;7(1):12115.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Firouzi S, Majid HA, Ismail A, Kamaruddin NA, Barakatun-Nisak MY. Effect of multi-strain probiotics (multi-strain microbial cell preparation) on glycemic control and other diabetes-related outcomes in people with type 2 diabetes: a randomized controlled trial. Eur J Nutr. 2017;56(4):1535–50.PubMedCrossRef Firouzi S, Majid HA, Ismail A, Kamaruddin NA, Barakatun-Nisak MY. Effect of multi-strain probiotics (multi-strain microbial cell preparation) on glycemic control and other diabetes-related outcomes in people with type 2 diabetes: a randomized controlled trial. Eur J Nutr. 2017;56(4):1535–50.PubMedCrossRef
30.
Zurück zum Zitat Jung SJ, Park SH, Choi EK, et al. Beneficial effects of Korean traditional diets in hypertensive and type 2 diabetic patients. J Med Food. 2014;17(1):161–71.PubMedPubMedCentralCrossRef Jung SJ, Park SH, Choi EK, et al. Beneficial effects of Korean traditional diets in hypertensive and type 2 diabetic patients. J Med Food. 2014;17(1):161–71.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Bayat A, Azizi-Soleiman F, Heidari-Beni M, et al. Effect of Cucurbita ficifolia and probiotic yogurt consumption on blood glucose, lipid profile, and inflammatory marker in type 2 diabetes. Int J Prev Med. 2016;7:30.PubMedPubMedCentralCrossRef Bayat A, Azizi-Soleiman F, Heidari-Beni M, et al. Effect of Cucurbita ficifolia and probiotic yogurt consumption on blood glucose, lipid profile, and inflammatory marker in type 2 diabetes. Int J Prev Med. 2016;7:30.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Feizollahzadeh S, Ghiasvand R, Rezaei A, Khanahmad H, Sadeghi A, Hariri M. Effect of probiotic soy milk on serum levels of adiponectin, inflammatory mediators, lipid profile, and fasting blood glucose among patients with type II diabetes mellitus. Probiotics Antimicrob Proteins. 2017;9(1):41–7.PubMedCrossRef Feizollahzadeh S, Ghiasvand R, Rezaei A, Khanahmad H, Sadeghi A, Hariri M. Effect of probiotic soy milk on serum levels of adiponectin, inflammatory mediators, lipid profile, and fasting blood glucose among patients with type II diabetes mellitus. Probiotics Antimicrob Proteins. 2017;9(1):41–7.PubMedCrossRef
33.
Zurück zum Zitat Khalili L, Alipour B, Asghari Jafar-Abadi M, et al. The effects of Lactobacillus casei on glycemic response, serum sirtuin1 and fetuin-a levels in patients with type 2 diabetes mellitus: a randomized controlled trial. Iran Biomed J. 2019;23(1):68–77.PubMedPubMedCentralCrossRef Khalili L, Alipour B, Asghari Jafar-Abadi M, et al. The effects of Lactobacillus casei on glycemic response, serum sirtuin1 and fetuin-a levels in patients with type 2 diabetes mellitus: a randomized controlled trial. Iran Biomed J. 2019;23(1):68–77.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Mohamadshahi M, Veissi M, Haidari F, Shahbazian H, Kaydani GA, Mohammadi F. Effects of probiotic yogurt consumption on inflammatory biomarkers in patients with type 2 diabetes. Bioimpacts. 2014;4(2):83–8.PubMedPubMedCentral Mohamadshahi M, Veissi M, Haidari F, Shahbazian H, Kaydani GA, Mohammadi F. Effects of probiotic yogurt consumption on inflammatory biomarkers in patients with type 2 diabetes. Bioimpacts. 2014;4(2):83–8.PubMedPubMedCentral
35.
Zurück zum Zitat Shakeri H, Hadaegh H, Abedi F, et al. Consumption of synbiotic bread decreases triacylglycerol and VLDL levels while increasing HDL levels in serum from patients with type-2 diabetes. Lipids. 2014;49(7):695–701.PubMedCrossRef Shakeri H, Hadaegh H, Abedi F, et al. Consumption of synbiotic bread decreases triacylglycerol and VLDL levels while increasing HDL levels in serum from patients with type-2 diabetes. Lipids. 2014;49(7):695–701.PubMedCrossRef
36.
Zurück zum Zitat Tajadadi-Ebrahimi M, Bahmani F, Shakeri H, et al. Effects of daily consumption of synbiotic bread on insulin metabolism and serum high-sensitivity C-reactive protein among diabetic patients: a double-blind, randomized, controlled clinical trial. Ann Nutr Metab. 2014;65(1):34–41.PubMedCrossRef Tajadadi-Ebrahimi M, Bahmani F, Shakeri H, et al. Effects of daily consumption of synbiotic bread on insulin metabolism and serum high-sensitivity C-reactive protein among diabetic patients: a double-blind, randomized, controlled clinical trial. Ann Nutr Metab. 2014;65(1):34–41.PubMedCrossRef
37.
Zurück zum Zitat Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, Niafar M, Asghari-Jafarabadi M, Mofid V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition. 2012;28(5):539–43.PubMedCrossRef Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, Niafar M, Asghari-Jafarabadi M, Mofid V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition. 2012;28(5):539–43.PubMedCrossRef
38.
Zurück zum Zitat Mazloom Z, Yousefinejad A, Dabbaghmanesh MH. Effect of probiotics on lipid profile, glycemic control, insulin action, oxidative stress, and inflammatory markers in patients with type 2 diabetes: a clinical trial. Iran J Med Sci. 2013;38(1):38–43.PubMedPubMedCentral Mazloom Z, Yousefinejad A, Dabbaghmanesh MH. Effect of probiotics on lipid profile, glycemic control, insulin action, oxidative stress, and inflammatory markers in patients with type 2 diabetes: a clinical trial. Iran J Med Sci. 2013;38(1):38–43.PubMedPubMedCentral
39.
Zurück zum Zitat Raygan F, Rezavandi Z, Bahmani F, et al. The effects of probiotic supplementation on metabolic status in type 2 diabetic patients with coronary heart disease. Diabetol Metab Syndr. 2018;10:51.PubMedPubMedCentralCrossRef Raygan F, Rezavandi Z, Bahmani F, et al. The effects of probiotic supplementation on metabolic status in type 2 diabetic patients with coronary heart disease. Diabetol Metab Syndr. 2018;10:51.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Hosseinzadeh P, Javanbakht MH, Mostafavi SA, et al. Brewer’s yeast improves glycemic indices in type 2 diabetes mellitus. Int J Prev Med. 2013;4(10):1131–8.PubMedPubMedCentral Hosseinzadeh P, Javanbakht MH, Mostafavi SA, et al. Brewer’s yeast improves glycemic indices in type 2 diabetes mellitus. Int J Prev Med. 2013;4(10):1131–8.PubMedPubMedCentral
41.
Zurück zum Zitat Kobyliak N, Falalyeyeva T, Mykhalchyshyn G, Kyriienko D, Komissarenko I. Effect of alive probiotic on insulin resistance in type 2 diabetes patients: randomized clinical trial. Diabetes Metab Syndr. 2018;12(5):617–24.PubMedCrossRef Kobyliak N, Falalyeyeva T, Mykhalchyshyn G, Kyriienko D, Komissarenko I. Effect of alive probiotic on insulin resistance in type 2 diabetes patients: randomized clinical trial. Diabetes Metab Syndr. 2018;12(5):617–24.PubMedCrossRef
42.
Zurück zum Zitat Mobini R, Tremaroli V, Ståhlman M, et al. Metabolic effects of Lactobacillus reuteri DSM 17938 in people with type 2 diabetes: a randomized controlled trial. Diabetes Obes Metab. 2017;19(4):579–89.PubMedCrossRef Mobini R, Tremaroli V, Ståhlman M, et al. Metabolic effects of Lactobacillus reuteri DSM 17938 in people with type 2 diabetes: a randomized controlled trial. Diabetes Obes Metab. 2017;19(4):579–89.PubMedCrossRef
43.
Zurück zum Zitat Sabico S, Al-Mashharawi A, Al-Daghri NM, et al. Effects of a 6-month multi-strain probiotics supplementation in endotoxemic, inflammatory and cardiometabolic status of T2DM patients: a randomized, double-blind, placebo-controlled trial. Clin Nutr. 2019;38(4):1561–9.PubMedCrossRef Sabico S, Al-Mashharawi A, Al-Daghri NM, et al. Effects of a 6-month multi-strain probiotics supplementation in endotoxemic, inflammatory and cardiometabolic status of T2DM patients: a randomized, double-blind, placebo-controlled trial. Clin Nutr. 2019;38(4):1561–9.PubMedCrossRef
44.
Zurück zum Zitat Ismail A, Darwish O, Tayel D, Elneily D, Elshaarawy G. Impact of probiotic intake on the glycemic control, lipid profile and inflammatory markers among patients with type 2 diabetes mellitus. Clin Diabetol. 2021;10(6):468–75. Ismail A, Darwish O, Tayel D, Elneily D, Elshaarawy G. Impact of probiotic intake on the glycemic control, lipid profile and inflammatory markers among patients with type 2 diabetes mellitus. Clin Diabetol. 2021;10(6):468–75.
45.
Zurück zum Zitat Hove KD, Brøns C, Færch K, Lund SS, Rossing P, Vaag A. Effects of 12 weeks of treatment with fermented milk on blood pressure, glucose metabolism and markers of cardiovascular risk in patients with type 2 diabetes: a randomised double-blind placebo-controlled study. Eur J Endocrinol. 2015;172(1):11–20.PubMedCrossRef Hove KD, Brøns C, Færch K, Lund SS, Rossing P, Vaag A. Effects of 12 weeks of treatment with fermented milk on blood pressure, glucose metabolism and markers of cardiovascular risk in patients with type 2 diabetes: a randomised double-blind placebo-controlled study. Eur J Endocrinol. 2015;172(1):11–20.PubMedCrossRef
46.
Zurück zum Zitat Palacios T, Vitetta L, Coulson S, et al. Targeting the intestinal microbiota to prevent type 2 diabetes and enhance the effect of metformin on glycaemia: a randomised controlled pilot study. Nutrients. 2020;12(7):2041.PubMedPubMedCentralCrossRef Palacios T, Vitetta L, Coulson S, et al. Targeting the intestinal microbiota to prevent type 2 diabetes and enhance the effect of metformin on glycaemia: a randomised controlled pilot study. Nutrients. 2020;12(7):2041.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Tonucci LB, Olbrich Dos Santos KM, Licursi de Oliveira L, Rocha Ribeiro SM, Duarte Martino HS. Clinical application of probiotics in type 2 diabetes mellitus: a randomized, double-blind, placebo-controlled study. Clin Nutr (Edinburgh, Scotland). 2017;36(1):85–92.CrossRef Tonucci LB, Olbrich Dos Santos KM, Licursi de Oliveira L, Rocha Ribeiro SM, Duarte Martino HS. Clinical application of probiotics in type 2 diabetes mellitus: a randomized, double-blind, placebo-controlled study. Clin Nutr (Edinburgh, Scotland). 2017;36(1):85–92.CrossRef
48.
Zurück zum Zitat Ostadrahimi A, Taghizadeh A, Mobasseri M, et al. Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: a randomized double-blind placebo-controlled clinical trial. Iran J Public Health. 2015;44(2):228–37.PubMedPubMedCentral Ostadrahimi A, Taghizadeh A, Mobasseri M, et al. Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: a randomized double-blind placebo-controlled clinical trial. Iran J Public Health. 2015;44(2):228–37.PubMedPubMedCentral
49.
Zurück zum Zitat Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism and personalized health care. Nat Rev Microbiol. 2005;3(5):431–8.PubMedCrossRef Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism and personalized health care. Nat Rev Microbiol. 2005;3(5):431–8.PubMedCrossRef
50.
Zurück zum Zitat Nicholson JK, Wilson ID. Opinion: understanding ‘global’ systems biology: metabonomics and the continuum of metabolism. Nat Rev Drug Discov. 2003;2(8):668–76.PubMedCrossRef Nicholson JK, Wilson ID. Opinion: understanding ‘global’ systems biology: metabonomics and the continuum of metabolism. Nat Rev Drug Discov. 2003;2(8):668–76.PubMedCrossRef
51.
Zurück zum Zitat Neves AL, Chilloux J, Sarafian MH, Rahim MB, Boulangé CL, Dumas ME. The microbiome and its pharmacological targets: therapeutic avenues in cardiometabolic diseases. Curr Opin Pharmacol. 2015;25:36–44.PubMedCrossRef Neves AL, Chilloux J, Sarafian MH, Rahim MB, Boulangé CL, Dumas ME. The microbiome and its pharmacological targets: therapeutic avenues in cardiometabolic diseases. Curr Opin Pharmacol. 2015;25:36–44.PubMedCrossRef
52.
Zurück zum Zitat Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55–60.PubMedCrossRef Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55–60.PubMedCrossRef
53.
Zurück zum Zitat Johnson EL, Heaver SL, Walters WA, Ley RE. Microbiome and metabolic disease: revisiting the bacterial phylum Bacteroidetes. J Mol Med (Berl). 2017;95(1):1–8.PubMedCrossRef Johnson EL, Heaver SL, Walters WA, Ley RE. Microbiome and metabolic disease: revisiting the bacterial phylum Bacteroidetes. J Mol Med (Berl). 2017;95(1):1–8.PubMedCrossRef
54.
Zurück zum Zitat Jonsson AL, Bäckhed F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 2017;14(2):79–87.PubMedCrossRef Jonsson AL, Bäckhed F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 2017;14(2):79–87.PubMedCrossRef
55.
Zurück zum Zitat Wang Y, Dilidaxi D, Wu Y, Sailike J, Sun X, Nabi XH. Composite probiotics alleviate type 2 diabetes by regulating intestinal microbiota and inducing GLP-1 secretion in db/db mice. Biomed Pharmacother. 2020;125: 109914.PubMedCrossRef Wang Y, Dilidaxi D, Wu Y, Sailike J, Sun X, Nabi XH. Composite probiotics alleviate type 2 diabetes by regulating intestinal microbiota and inducing GLP-1 secretion in db/db mice. Biomed Pharmacother. 2020;125: 109914.PubMedCrossRef
56.
Zurück zum Zitat Pizarroso NA, Fuciños P, Gonçalves C, Pastrana L, Amado IR. A Review on the Role of Food-Derived Bioactive Molecules and the Microbiota-Gut-Brain Axis in Satiety Regulation. Nutrients. 2021;13(2):632.PubMedPubMedCentralCrossRef Pizarroso NA, Fuciños P, Gonçalves C, Pastrana L, Amado IR. A Review on the Role of Food-Derived Bioactive Molecules and the Microbiota-Gut-Brain Axis in Satiety Regulation. Nutrients. 2021;13(2):632.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Martin-Gallausiaux C, Marinelli L, Blottière HM, Larraufie P, Lapaque N. SCFA: mechanisms and functional importance in the gut. Proc Nutr Soc. 2021;80(1):37–49.PubMedCrossRef Martin-Gallausiaux C, Marinelli L, Blottière HM, Larraufie P, Lapaque N. SCFA: mechanisms and functional importance in the gut. Proc Nutr Soc. 2021;80(1):37–49.PubMedCrossRef
58.
Zurück zum Zitat Mueller NT, Zhang M, Juraschek SP, Miller ER, Appel LJ. Effects of high-fiber diets enriched with carbohydrate, protein, or unsaturated fat on circulating short chain fatty acids: results from the OmniHeart randomized trial. Am J Clin Nutr. 2020;111(3):545–54.PubMedPubMedCentralCrossRef Mueller NT, Zhang M, Juraschek SP, Miller ER, Appel LJ. Effects of high-fiber diets enriched with carbohydrate, protein, or unsaturated fat on circulating short chain fatty acids: results from the OmniHeart randomized trial. Am J Clin Nutr. 2020;111(3):545–54.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol. 2019;16(8):461–78.PubMedCrossRef Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol. 2019;16(8):461–78.PubMedCrossRef
61.
Zurück zum Zitat Maldonado-Gómez MX, Martínez I, Bottacini F, et al. Stable engraftment of Bifidobacterium longum AH1206 in the human gut depends on individualized features of the resident microbiome. Cell Host Microbe. 2016;20(4):515–26.PubMedCrossRef Maldonado-Gómez MX, Martínez I, Bottacini F, et al. Stable engraftment of Bifidobacterium longum AH1206 in the human gut depends on individualized features of the resident microbiome. Cell Host Microbe. 2016;20(4):515–26.PubMedCrossRef
62.
Zurück zum Zitat Xiao Y, Zhao J, Zhang H, Zhai Q, Chen W. Mining Lactobacillus and Bifidobacterium for organisms with long-term gut colonization potential. Clin Nutr (Edinburgh, Scotland). 2020;39(5):1315–23.CrossRef Xiao Y, Zhao J, Zhang H, Zhai Q, Chen W. Mining Lactobacillus and Bifidobacterium for organisms with long-term gut colonization potential. Clin Nutr (Edinburgh, Scotland). 2020;39(5):1315–23.CrossRef
63.
Zurück zum Zitat Fiocco D, Longo A, Arena MP, Russo P, Spano G, Capozzi V. How probiotics face food stress: they get by with a little help. Crit Rev Food Sci Nutr. 2020;60(9):1552–80.PubMedCrossRef Fiocco D, Longo A, Arena MP, Russo P, Spano G, Capozzi V. How probiotics face food stress: they get by with a little help. Crit Rev Food Sci Nutr. 2020;60(9):1552–80.PubMedCrossRef
64.
Zurück zum Zitat Bove P, Gallone A, Russo P, et al. Probiotic features of Lactobacillus plantarum mutant strains. Appl Microbiol Biotechnol. 2012;96(2):431–41.PubMedCrossRef Bove P, Gallone A, Russo P, et al. Probiotic features of Lactobacillus plantarum mutant strains. Appl Microbiol Biotechnol. 2012;96(2):431–41.PubMedCrossRef
65.
Zurück zum Zitat Charteris WP, Kelly PM, Morelli L, Collins JK. Development and application of an in vitro methodology to determine the transit tolerance of potentially probiotic Lactobacillus and Bifidobacterium species in the upper human gastrointestinal tract. J Appl Microbiol. 1998;84(5):759–68.PubMedCrossRef Charteris WP, Kelly PM, Morelli L, Collins JK. Development and application of an in vitro methodology to determine the transit tolerance of potentially probiotic Lactobacillus and Bifidobacterium species in the upper human gastrointestinal tract. J Appl Microbiol. 1998;84(5):759–68.PubMedCrossRef
66.
Zurück zum Zitat Huang Y, Adams MC. In vitro assessment of the upper gastrointestinal tolerance of potential probiotic dairy propionibacteria. Int J Food Microbiol. 2004;91(3):253–60.PubMedCrossRef Huang Y, Adams MC. In vitro assessment of the upper gastrointestinal tolerance of potential probiotic dairy propionibacteria. Int J Food Microbiol. 2004;91(3):253–60.PubMedCrossRef
68.
Zurück zum Zitat Tiwari SK. Bacteriocin-producing probiotic lactic acid bacteria in controlling dysbiosis of the gut microbiota. Front Cell Infect Microbiol. 2022;12:851140.PubMedPubMedCentralCrossRef Tiwari SK. Bacteriocin-producing probiotic lactic acid bacteria in controlling dysbiosis of the gut microbiota. Front Cell Infect Microbiol. 2022;12:851140.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Hurkala J, Lauterbach R, Radziszewska R, Strus M, Heczko P. Effect of a short-time probiotic supplementation on the abundance of the main constituents of the gut microbiota of term newborns delivered by cesarean section: a randomized, prospective, controlled clinical trial. Nutrients. 2020;12(10):3128.PubMedPubMedCentralCrossRef Hurkala J, Lauterbach R, Radziszewska R, Strus M, Heczko P. Effect of a short-time probiotic supplementation on the abundance of the main constituents of the gut microbiota of term newborns delivered by cesarean section: a randomized, prospective, controlled clinical trial. Nutrients. 2020;12(10):3128.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Ibarra A, Latreille-Barbier M, Donazzolo Y, Pelletier X, Ouwehand AC. Effects of 28-day Bifidobacterium animalis subsp. lactis HN019 supplementation on colonic transit time and gastrointestinal symptoms in adults with functional constipation: a double-blind, randomized, placebo-controlled, and dose-ranging trial. Gut Microbes. 2018;9(3):236–51.PubMedPubMedCentralCrossRef Ibarra A, Latreille-Barbier M, Donazzolo Y, Pelletier X, Ouwehand AC. Effects of 28-day Bifidobacterium animalis subsp. lactis HN019 supplementation on colonic transit time and gastrointestinal symptoms in adults with functional constipation: a double-blind, randomized, placebo-controlled, and dose-ranging trial. Gut Microbes. 2018;9(3):236–51.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Whorwell PJ, Altringer L, Morel J, et al. Efficacy of an encapsulated probiotic Bifidobacterium infantis 35624 in women with irritable bowel syndrome. Am J Gastroenterol. 2006;101(7):1581–90.PubMedCrossRef Whorwell PJ, Altringer L, Morel J, et al. Efficacy of an encapsulated probiotic Bifidobacterium infantis 35624 in women with irritable bowel syndrome. Am J Gastroenterol. 2006;101(7):1581–90.PubMedCrossRef
Metadaten
Titel
The effects of probiotics supplementation on glycaemic control among adults with type 2 diabetes mellitus: a systematic review and meta-analysis of randomised clinical trials
verfasst von
Guang Li
Hao Feng
Xin-Liang Mao
Yan-Jun Deng
Xiao-Bao Wang
Qiong Zhang
Yan Guo
Su-Mei Xiao
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2023
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-023-04306-0

Weitere Artikel der Ausgabe 1/2023

Journal of Translational Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

RAS-Blocker bei Hyperkaliämie möglichst nicht sofort absetzen

14.05.2024 Hyperkaliämie Nachrichten

Bei ausgeprägter Nierenfunktionsstörung steigen unter der Einnahme von Renin-Angiotensin-System(RAS)-Hemmstoffen nicht selten die Serumkaliumspiegel. Was in diesem Fall zu tun ist, erklärte Prof. Jürgen Floege beim diesjährigen Allgemeinmedizin-Update-Seminar.

Gestationsdiabetes: In der zweiten Schwangerschaft folgenreicher als in der ersten

13.05.2024 Gestationsdiabetes Nachrichten

Das Risiko, nach einem Gestationsdiabetes einen Typ-2-Diabetes zu entwickeln, hängt nicht nur von der Zahl, sondern auch von der Reihenfolge der betroffenen Schwangerschaften ab.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Battle of Experts: Sport vs. Spritze bei Adipositas und Typ-2-Diabetes

11.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Im Battle of Experts traten zwei Experten auf dem Diabeteskongress gegeneinander an: Die eine vertrat die Auffassung „Sport statt Spritze“ bei Adipositas und Typ-2-Diabetes, der andere forderte „Spritze statt Sport!“ Am Ende waren sie sich aber einig: Die Kombination aus beidem erzielt die besten Ergebnisse.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.