Vous êtes sur la page 1sur 30

Accepted Manuscript

Clinical Application of Probiotics in Type 2 Diabetes Mellitus: a Randomized, Double-


Blind, Placebo-Controlled Study

Livia Bordalo Tonucci, PhD, Professor, Karina Maria Olbrich dos Santos, PhD,
Leandro Licursi de Oliveira, PhD, Sonia Machado Rocha Ribeiro, PhD, Hercia
Stampini Duarte Martino, PhD
PII: S0261-5614(15)00331-3
DOI: 10.1016/j.clnu.2015.11.011
Reference: YCLNU 2692

To appear in: Clinical Nutrition

Received Date: 12 May 2015


Revised Date: 8 November 2015
Accepted Date: 12 November 2015

Please cite this article as: Tonucci LB, dos Santos KMO, Oliveira LLd, Ribeiro SMR, Martino HSD,
Clinical Application of Probiotics in Type 2 Diabetes Mellitus: a Randomized, Double-Blind, Placebo-
Controlled Study, Clinical Nutrition (2016), doi: 10.1016/j.clnu.2015.11.011.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
1
ACCEPTED MANUSCRIPT
Clinical Application of Probiotics in Type 2 Diabetes Mellitus: a Randomized,

Double-Blind, Placebo-Controlled Study

Authors:

Livia Bordalo Tonuccia, PhD

Karina Maria Olbrich dos Santosb, PhD

PT
Leandro Licursi de Oliveirac, PhD

Sonia Machado Rocha Ribeirod, PhD

RI
Hercia Stampini Duarte Martinod, PhD

SC
a
Professor at Department of Nutrition, INTA College.
b

U
Brazilian Agricultural Research Corporation (Embrapa). Address: Fazenda Três
AN
Lagoas, Estrada Sobral-Groaíras, Km 4. Postal Code 145. Zip Code 62011-970.
c
M

Professor at Department of Biology, Federal University of Viçosa. Address: Av. PH

Rolfs, s/n, Campus UVF, CCB II – Viçosa, MG, Brazil. Zip Code: 36.570-000.
D

d
Professor at Department of Nutrition and Health, Federal University of Viçosa.
TE

Address: Av. PH Rolfs, s/n, Campus UVF, CCB II – Viçosa, MG, Brazil. Zip Code:

36.570-000.
EP

Corresponding author: Livia Bordalo Tonucci. Address: R. Antônio Rodrigues


C

Magalhães, 359 - Dom Expedito Lopes, Sobral - CE. Zip Code: 62050-100. Postal
AC

code, 10. E-mail: livia_bordalo@hotmail.com. Phones: 55 (88) 98809-9885/ Fax

number: (31) 3899-3176.

Short running head: Clinical Application of Probiotics in Diabetes

Sources of support: EMBRAPA and FAPEMIG, Brazil


Clinical trial registry: This trial is registered with ensaiosclinicos.gov.br/rg/RBR-
219644.
1
ACCEPTED MANUSCRIPT
1 ABSTRACT

2 Background & Aims: Type 2 diabetes has been associated with dysbiosis and one of the

3 possible routes to restore a healthy gut microbiota is by the regular ingestion of probiotics.

4 We aimed to investigate the effects of probiotics on glycemic control, lipid profile,

5 inflammation, oxidative stress and short chain fatty acids in T2D.

PT
6 Methods: In a double-blind, randomized, placebo-controlled trial, 50 volunteers consumed

RI
7 daily 120 g/d of fermented milk for 6 wk. Participants were assigned into two groups:

8 probiotic group, consuming fermented milk containing Lactobacillus acidophilus La-5 and

SC
9 Bifidobacterium animalis subsp lactis BB-12 (109 colony-forming units/d, each) and control

10 group, consuming conventional fermented milk. Anthropometric measurements, body

11
U
composition, fasting blood and faecal samples were taken at baseline and after 6 wk.
AN
12 Results: 45 subjects out of 50 (90%) completed follow-up. After 6 wk, there was a
M

13 significant decrease in fructosamine levels (- 9.91mmol/L; p = 0.04) and hemoglobin A1c

14 tended to be lower (- 0.67%; p = 0.06) in probiotic group. TNF-α and resistin were
D

15 significantly reduced in probiotic and control groups (- 1.5 and -1.3 pg/mL, -2.1 and -2.8
TE

16 ng/mL, respectively), while IL-10 was significantly reduced (- 0.65 pg/mL; p < 0.001) only

17 in the control group. Fecal acetic acid was increased in both groups (0.58 and 0.59% in
EP

18 probiotic and control groups, respectively; p < 0.01). There was a significant difference
C

19 between groups concerning mean changes of HbA1c (+ 0.31 for control group vs - 0.65 for
AC

20 probiotic group; p = 0.02), total cholesterol (+ 0.55 for control group vs - 0.15 for probiotic

21 group; p = 0.04) and LDL-cholesterol (+ 0.36 for control group vs - 0.20 for probiotic group

22 p = 0.03).

23 Conclusions: Probiotic consumption improved the glycemic control in T2D subjects,

24 however, the intake of fermented milk seems to be involved with others metabolic changes,

25 such as decrease in inflammatory cytokines (TNF-α and resistin) and increase in the acetic
2
ACCEPTED MANUSCRIPT
26 acid.

27 1. Introduction

28 Diabetes mellitus is on the rise all over the world affecting more than 382 million

29 people. Type 2 diabetes (T2D) accounts for 85% to 95% of all diabetes and is a complex

30 chronic illness requiring multifactorial risk reduction strategies [1].

PT
31 T2D is often associated with systemic inflammation [2] and increased oxidative stress

RI
32 [3]. Both molecular and metabolic mechanisms are links to β-cell dysfunction and/or insulin

33 resistance [4]. However, the intake of probiotics has been demonstrated to reduce

SC
34 inflammation and oxidative stress markers, and to improve glycemic and insulin metabolism

35 [5-6].

36
U
In addition, the gut microbiota also seems to be important to the pathophysiology of
AN
37 T2D [7]. Findings from two studies that used faecal samples suggested that compositional
M

38 and functional changes in the gut microbiota might be directly linked to the development of

39 T2D [8-9]. Various others mechanisms have been proposed to explain the influence of the
D

40 microbiota on insulin resistance and T2D, such as metabolic endotoxemia [10], modifications
TE

41 in the secretion of the incretins [11], and short-chain fatty acids (SCFA) production [12].

42 Multiple cytokines are involved in the development of T2D, and are fundamental signals in
EP

43 the intestinal immune system, able to subvert the physiological status of inflammation in the
C

44 gut [13].
AC

45 Alterations in the gut microbiota as a result of probiotics and prebiotics intake have

46 been reported. Experimental studies have reported beneficial effects by strains of

47 Lactobacillus on glycemic control, stress oxidative and/or inflammation in animal model of

48 type 2 diabetes [5, 14]. However, the effects of probiotics on diabetes endpoints remain

49 unknown.

50 The objective of this study was to investigate the efficacy of the intake of fermented
3
ACCEPTED MANUSCRIPT
51 goat milk containing Lactobacillus acidophilus La-5 and Bifidobacterium animalis BB-12 on

52 glycemic control, lipid profile, inflammation, oxidative stress and faecal SCFA in T2D

53 subjects.

54

55 2. Methods

PT
56 2.1 Subjects

RI
57 This randomised, double-blind, parallel-group, placebo-controlled trial was carried

58 out in Ceará, Brazil, from June 2013 to February 2014. The study was performed on 50

SC
59 volunteers with T2D according to ADA criteria [15], age 35–60 y, body mass index (BMI)

60 lower than 35 kg/m2 and type 2 diabetes diagnosed for at least one year, recruited from two

61
U
endocrinology clinics. The appropriate sample size was calculated based on primary outcome
AN
62 of serum interleukin (IL)-6 levels obtained from Mazloom et al. [16], considering a alpha
M

63 level of 0.05 and a power of 80%. The number of subjects required was 22.5 per group, but

64 this number was increased to 25 per group to accommodate the withdrawals. The research
D

65 team did recruitment within the clinics after indication of participants by endocrinologist.
TE

66 We excluded participants with clinical evidence of chronic illness or gastrointestinal

67 disorders; presence of renal, hepatic, haematological or immunodeficiency diseases; history


EP

68 of cancer or cardiovascular diseases; current treatment with insulin, DPP-4 inhibitor or GLP-
C

69 1 analogue; smoking; any intake of probiotics, supplements, antiobesity and anti-


AC

70 inflammatory drugs, and antibiotics in the three months preceding recruitment, and

71 pregnancy or breast-feeding.

72 The study protocol was approved by the Federal University of Viçosa Ethics

73 Committee. All patients provided written informed consent. This trial is registered with

74 ensaiosclinicos.gov.br/rg/RBR-219644.

75 2.2 Randomisation and blinding


4
ACCEPTED MANUSCRIPT
76 Participants were randomly assigned (1:1) by computer (Research Randomizer,

77 version 4.0) to either probiotic or control group in block sizes of four. The randomization was

78 stratified by gender. Volunteers, investigators, and trial staff were masked to treatment

79 allocation. However, investigators analysing study data were not masked. Blinding was done

80 by a technical assistant, right after beverages production.

PT
81 2.3 Study protocol

RI
82 T2D subjects were randomly assigned to consume either 120 g/d of probiotic

83 fermented goat milk containing 109 UFCs of Lactobacillus acidophilus La-5 and 109 UFCs of

SC
84 Bifidobacterium animalis subsp. lactis BB-12 (probiotic group) or 120 g/d of conventional

85 fermented goat milk contained Streptococcus thermophilus TA-40 (control group) for 6 wk,

86
U
during the breakfast. The fermented milk (FM) was delivered every 2 wk and volunteers were
AN
87 instructed to keep it under refrigeration (4 ºC).
M

88 The probiotic and conventional FM were developed in parthership with Embrapa Goat

89 and Sheep (Ceará, Brazil). Pasteurised milk was cooled to 43 ± 2oC for the addition of the
D

90 starter culture (S. thermophilus TA-40; Danisco, Sassenage, France), and the probiotic
TE

91 cultures (L. acidophilus La-5 and B. animalis subsp. lactis BB-12; Chr. Hansen, Hoersholm,

92 Denmark). The beverages were flavored with grape juice obtained from Embrapa Grape and
EP

93 Wine (Rio Grande do Sul, Brazil). Ingredients, chemical composition and antioxidants status
C

94 of the probiotic FM are shown in Table supplementary 1 (see supplementary data). Both
AC

95 products had the same taste, color, and smell.

96 Fermented milk was sampled immediately after manufacture and delivered on the

97 following day. Microbiological analysis was conducted every week during the trial. S.

98 thermophilus enumeration was performed on M17 agar, containing lactose (Vetec, Duque de

99 Caxias, Brazil, 5 g/L) and incubated aerobically at 37 ºC for 48 h. B. lactis and L. acidophilus

100 were determined in modified De Man Rogosa Sharpe (MRS) agar (Oxoid, Basingstoke, UK),
5
ACCEPTED MANUSCRIPT
101 followed by incubation at 37 ºC for 72 h [17].

102 Microbiological analyses of the probiotic FM showed that the average colony counts

103 of probiotic bacteria on days 1, 7 and 14 were 7.72 x 107, 5.82 x 107, and 1.62 x 106 CFUs/g

104 of L. acidophilus La-5 and 4.45 x 108, 1.84 x 107, and 1.56 x 107CFUs/g of B. lactis BB-12,

105 respectively. Both probiotic bacteria showed an appropiate survival rate during 14 days

PT
106 storage time (see Supplemental Table 2). Additionaly, sensory evaluation was carried out in

RI
107 FM samples during the trial through acceptability tests, using the hybrid hedonic scale (1 =

108 disliked extremely, 5 = neither liked nor disliked, 09 = liked extremely) [18]. The overall

SC
109 acceptability of the probiotic and conventional FM was 8.17 and 8.20, respectively (see

110 Supplemental Table 3).

111
U
All anthropometric and biochemical measures were conducted in a fasting state taken
AN
112 at baseline and after 6 wk intervention. A single experienced examiner performed all
M

113 anthropometric measurements. Body weight and body composition were assessed by bio-

114 impedance (Tanita TBF-300, Tanita Corporation, Tokyo, Japan). Waist circumference
D

115 (measured midway between lowest rib and iliac crest) was measured using a non-stretchable
TE

116 measuring tape (Sanny, São Paulo, Brazil).

117 Dietary assessment was done using food records, which were conducted by the
EP

118 nutritionist in the first and the last week of intervention. Dietary intake was analysed using
C

119 Avanutri Revolution software (Rio de Janeiro, Brazil). Regular level of physical activity was
AC

120 defined as three times a week for at least 30 min.

121 Subjects were instructed to follow their usual diets, level of physical activity, and

122 lifestyle, avoiding any changes in medication, and unusual or excessive food and alcohol

123 drink consumption throughout the intervention period. We contacted the volunteers once a

124 week to ask for adverse health events and to assess the adherence to treatment.

125 2.4 Biochemical measurements


6
ACCEPTED MANUSCRIPT
126 For the biochemical measurements, blood samples were obtained after a 12 h

127 overnight fast. Before the test day, the subjects were instructed not to consume alcohol and to

128 refrain from heavy physical activity during 72 and 24 h, respectively. Samples were

129 centrifuged at 1,000 g for 10 min at 4 °C, aliquoted and analysed or immediately stored at –

130 80 °C for cytokines and oxidative stress analyses.

PT
131 Blood samples were analyzed for fasting plasma glucose (FPG) and lipid profile by

RI
132 enzymatic colorimetric method (Bioclin, Minas Gerais, Brazil, and Beckman Coulter,

133 California, USA). Insulin was measure through electrochemiluminescence (Modular

SC
134 AnalyticsDxI800, Beckman Coulter, California, USA). Fructosamine was assayed by

135 colorimetric method (BioSystems, Barcelona, Spain), and HbA1c with ion exchange high-

136
U
performance liquid chromatography (Bio-rad kit, California, USA). The homeostasis model
AN
137 assessment index (HOMA-IR) was used as an indicator of insulin resistance and calculated as
M

138 follows: HOMA-IR = fasting plasma insulin (µU/mL) x fasting plasma glucose (mmol/

139 L)/22.5. Insulin resistance was diagnosed using a cut off value of 2.71 [19].
D

140 For determination of oxidative stress markers, plasma samples were collected in
TE

141 vacutainers containing sodium citrate. Colorimetric assay (Sigma-Aldrich antioxidant assay

142 kit, Missouri, USA; interassay coefficients of variation 3%) was used to measure plasma total
EP

143 antioxidant status (TAS). Plasma total F2–isoprostane was measured by ELISA kit (Caymans
C

144 8-isoprostane EIA kit, Michigan, USA; interassay coefficients of variation 9.5% at 80 pg/mL
AC

145 and 6.4% at 32 pg/mL). Briefly, plasma samples (400 µL) were hydrolysed with (10N)

146 NaOH (100 µL) at 45 °C for 2 h to measure both free and esterified isoprostane. After

147 incubation, 100 µL of (10M) HCl was added and the samples were centrifuged at 1,500g for

148 10 min to remove precipitated proteins.

149 Cytokine concentrations were determined using a multiplexed bead immunoassay.

150 This is a bead-based suspension array using the LuminexxMAP technology in which
7
ACCEPTED MANUSCRIPT
151 fluorescent-coded beads, known as microspheres, have cytokine capture antibodies on the

152 bead surface to bind the proteins. The measures of 5 cytokines (IL-6, IL-10, TNF-α,

153 adiponectin, and resistin) were measured using the human Cytokine/Adipokine magnetic

154 bead kits (Millipore Coproration, Missouri, USA). Assays were performed in 96-well filter

155 plates, as previously described [20]. The concentration of the samples was estimated from the

PT
156 standard curve using a fifth-order polynomial equation (Software xPonent/Analyst versão

RI
157 4.2). Intra-assay coefficient of variation was 2.0, 1.6, 2.6, 4.0, and 3.0 % for IL-6, IL-10,

158 TNF-α, adiponectin, and resistin, respectively.

SC
159 2.5 Faecal analysis

160 Feces samples (5-10g) were collected at enrollment and at the sixth week. Samples

161
U
(blinded) were immediately placed at -80 °C and stored until analyzed. The extraction of
AN
162 SCFA (acetate, butyrate, and propionate) was based on the method of Smiricky-Tjardes et
M

163 al.[21] and measured by GC (model GC-17A, Shimadzuw, Maryland, USA). N2 was used as

164 the carrier gas and the flux in the column was 1.0 mL/min. The temperatures of the injector
D

165 and detector were set at 220 and 250°C, respectively. Initial column temperature was 100°C
TE

166 sustained for 5 min, rising at 108°C/min until it reached 185°C. Next, the samples were

167 injected (1 mL) through a Hamiltonw syringe (10 mL) in split system 5. The results were
EP

168 represented as per 100 mg of faeces (% w/w).


C

169 2.6 Statistical Analysis


AC

170 Statistical analysis was done using SPSS Statistics version 20 (IBM, NY, USA). All

171 data were checked for normal distribution using Shapiro-Wilk test and Skewness/Kurtosis.

172 Data were reported as mean (SD), and median and interquartile interval (P25 and P75%),

173 since some variables were not normally distributed (HbA1c, HOMA-IR, IL-6, and acetic

174 acid). Comparison between probiotic and control groups at baseline was tested using

175 unpaired Student’s t test or Mann-Whitney test. Paired Student’s t test or Wilcoxon matched-
8
ACCEPTED MANUSCRIPT
176 pairs signed-rank test were used to analyze differences between baseline and endpoint values.

177 Pearson or Spearman’s correlation tests were performed to measure the degree of correlation

178 between cytokine concentrations and glycemic control. A p value < 0.05 was considered

179 statistically significant.

180 3. Results

PT
181 A total of 45 (90 %) subjects aged 35 to 60 y (mean 51.40 ± 6.80) concluded this

RI
182 study. Five patients were excluded according to reasons shown in Figure 1. Abdominal

183 discomfort was the only reported adverse effect. This subject (n = 1) belonged to the control

SC
184 group and was withdrawn from the study.

185 Baseline characteristics are shown in Table 1. The main drugs used by the volunteers

186
U
were metformin (94% in the probiotic group and 95.5% in the control group) and
AN
187 glibenclamide (44% in the probiotic group and 41% in the control group). At the baseline, no
M

188 relevant differences could be detected in the general characteristics between control and

189 probiotics group, except for HbA1c (p = 0.04), which was higher in the probiotic group.
D

190 There were no statistically significant differences in anthropometric and body composition
TE

191 values between or within groups at the end of the study (data not shown). Similarly, at the

192 beginning of the study, no significant differences were found between the two groups in
EP

193 terms of dietary intakes. Comparing the dietary intakes throughout the study separately in
C

194 each group, we observed no significant differences within group (see Supplemental Table 4).
AC

195 3.1 Impact of the intervention on glicemic control

196 Biochemical markers after probiotic treatment are show in Table 2. At 6-week follow-

197 up, the consumption of probiotic fermented milk significantly decreased fructosamine levels

198 (-9.91 mmol/L; p = 0.04) and HbA1c levels tended to be reduced (-0.67%; p = 0.06), while in

199 the control group no significant effect was detected on glycemic control (p > 0.05). When the

200 median changes in HbA1c were compared between groups, there was a significant difference
9
ACCEPTED MANUSCRIPT
201 (+ 0.31 for control group vs - 0.65 for probiotic group; p = 0.02). The fasting plasma glucose

202 (FPG), insulin concentrations, as well as insulin resistance, evaluated by the HOMA index,

203 did not change significantly throughout the follow-up period in both groups (p > 0.05 for all).

204 3.2 Effect of probiotics on lipid profile

205 Within group comparisons of lipid profile revealed that consumption of probiotic

PT
206 fermented milk prevented a rise in total cholesterol (TC) and LDL-C, while in the control

RI
207 group we observed a significant increase in the TC and LDL-c (11.35 and 16.10%; P =0.01

208 and P =0.04, respectively). Furthermore, when the mean changes were compared between

SC
209 the two groups, there was a statistically significant difference in the TC (- 0.58mmol/L, 95%

210 CI: – 1.13 to – 0.03; p = 0.04) and LDL-C (-0.20 mmol/L, 95% CI: -1.02 to -0.03; p = 0.03).

211
U
At the end of trial, no significant effect on HDL-C, VLDL, and triglycerides were found in
AN
212 both groups (p > 0.05 for all). The TC:HDL-C ratio was significantly increased by 8.94%
M

213 (0.28 mmol/L) in the control group during the study (p =0.03), while no statistically

214 significant changes (p = 0.75) was reported in probiotic group (Table 2).
D

215 3.3 Effect of probiotics on markers of oxidative stress


TE

216 At baseline, there was no difference between groups in TAS (0.01mM; 95% CI: –

217 0.06 to 0.07; p = 0.86) and F2-isoprostane levels (11.66 pg/dL; 95% CI: – 8.77 to 32.11; p
EP

218 =0.25). No significant difference was detected in plasma TAS concentrations from baseline
C

219 to post intervention in probiotic or control groups (p =0.40 and P =0.23, respectively) (Table
AC

220 2). Similarly, plasma F2-isoprostane concentrations did not change in the probiotic (p = 0.76)

221 or control (p = 0.94) group over time.

222 3.4 Effect of probiotics on cytokines levels

223 Anti-inflammatory markers, such as IL-10 and adiponectin, decreased after

224 intervention in control group (- 0.64 pg/mL [95% CI: -0.47 to -0.81; p = 0.001] for IL-10, -

225 0.76 µg/mL [95% CI: -0.75 to -1.59; p = 0.07] for adiponectin), while no significant change
10
ACCEPTED MANUSCRIPT
226 was observed in the probiotic group (p = 0.38 and p = 0.14, respectively). At the end of trial,

227 the two groups exhibited reduction in TNF- α (- 1.4 pg/mL [95% CI: - 0.01 to 2.79; p =0.04]

228 for probiotic, -1.36 pg/mL [95% CI: -0.24 to -2.5; P =0.02] for control group) and resistin (-

229 2.06 ng/mL [95% CI: -0.66 to -3.44; p = 0.006] for probiotic, - 2.80ng/mL [95% CI: -1.45 to

230 - 4.16; p =0.001] for control group). No significant differences in IL-6 levels were observed

PT
231 in any groups post intervention. Reductions in resistin were positively correlated with

RI
232 reductions in HbA1c (r = 0.320; p = 0.03). No correlation was found between other cytokines

233 and markers of glycemic control (data not shown). In Fig. 1 it is possible to compare the

SC
234 cytokines concentration between the two groups .

235 3.5 Effect of probiotics on faecal SCFA analysis

236
U
Data from faecal analysis showed a significant increase in the acetic acid in both
AN
237 groups (0.58 % [95% CI: 0.97 to 1.96; p = 0.005] for probiotic, 0.59 % [95% CI: 0.98 to
M

238 1.89; p = 0.006] for control group) at the end of trial. However, there were no significant

239 differences in the butyric and propionic acids after intervention in control and probiotic
D

240 groups, as shown in Fig. 2. Additionally, no significant difference was found between
TE

241 changes of the two groups in butyric, acetic and propionic acids (p > 0.05). Interestingly, a

242 higher proportion of propionic acid and a lower proportion of butyric acid were recorded in
EP

243 both groups. At end of trial, the proportion of propionic: acetic: butyric acids, taking into
C

244 account the mean values, was also similar: 10: 8: 1 in the control group and 14: 10: 1 in the
AC

245 probiotic group.

246

247 4. Discussion

248 This is the first clinical trial to assess the impact of probiotic use on fructosamine,

249 faecal SCFA, IL-10, resistin, adiponectin, and F2-isoprostane in T2D patients. It is speculated

250 that improvement in the inflammatory markers, stress oxidative status, and SCFA contributes
11
ACCEPTED MANUSCRIPT
251 to diabetes control [14, 22].

252 The present study showed that fermented milk consumption significantly decreased

253 inflammatory cytokines (TNF-α and resistin) in both treatments (control vs probiotic), as well

254 as caused a significant increase in the acetic acid, but only the probiotic group showed

255 improvement in glycemic control, as demonstrated by fructosamine and HbA1c. The main

PT
256 differences between the results were: significant decrease in IL-10, a tendence of decreasing

RI
257 in adiponectin levels, and significant increase in TC and LDL-C, which were observed only

258 in control group. These results indicate that these last factors hinder the improvement in

SC
259 glycemic control of subjects in the control group. Furthermore, total phenolic content and

260 antioxidant activity were significantly higher in the probiotic fermented milk (see

261 Supplemental Table 1).


U
AN
262 Most of the previous animal studies that evaluated the effect of probiotics on glycemic
M

263 control in T2D, related that probiotics, especially Lactobacillus, can reduced FPG, HbA1c,

264 and insulin, beyond inflammatory markers, such as IL-2, INF-γ, IL-6, and TNF-α [5, 23-24].
D

265 The intervention time ranged from 8 to 14 wk. Increase in GLUT4 mRNA expression levels
TE

266 in adipose tissue has also been reported [5, 25].

267 Concerning clinical trials, previous studies including diabetic subjects have shown
EP

268 controversial results, especially regarding glycemic control and oxidative stress. Ejtahed et

al., [26] reported reductions in HbA1c (p < 0.05) after 6 wk of yogurt intake containing 1010
C

269
AC

270 CFUs of L. acidophilus La-5 and B. lactis BB-12. On the other hand, L. acidophilus NCFM

271 capsule (1010 CFUs) intake during 4 wk, did not change the glycemic control, insulin, IL-1,

272 IL-6, TNF- α, and protein C reactive (CRP) in T2D subjects [27]. Also, multispecies

273 probiotic supplementation (L. acidophilus, L. casei, L. rhamnosus, L. bulgaricus, B. breve, B.

274 longum, and Streptococcus thermophilus - 109 CFUs each) associated with fructo-

275 oligosaccharide (100 mg) during 8 wk did not result in significant reductions in FPG and a
12
ACCEPTED MANUSCRIPT
276 significant increase in the levels of insulin, HOMA-IR, and LDL-C was found in both groups

277 [28]. Subsequently, this same research group reported that daily consumption of Bacillus

278 coagulans (107 CFUs) and 1.08g of inulin during 6 wk, did not result in changes in FPG and

279 HOMA-IR (p > 0.05), although a significant decrease in hs-CRP and a significant increase in

280 glutathione peroxidase (GPx) activity were found [29].

PT
281 The measurement of plasma F2-isoprostane is considered the best measure of plasma

RI
282 oxidative stress and TAS gives the total sum of both exogenous and endogenous antioxidants

283 [30] showing the complete antioxidant status picture. It has been suggested that the effects of

SC
284 probiotic on glycemic control can be partly explained by the effects on oxidative stress [31].

285 However, the present study did not show a significant impact of probiotics on F2-isoprostane

286
U
or TAS plasma levels, as well as, earlier clinical trial that evaluated TAS after 6 wk intake of
AN
287 B. coagulans (107 CFUs) and 1g of inulin in 124 diabetic patients [29]. On the order hand,
M

288 after consumption of probiotic yogurt containing L. acidophilus La-5 and B. lactis BB-12 for

289 6 wk, there was an increase in erythrocyte superoxide dismutase (SOD) and GPx activities,
D

290 and TAS (p < 0.05), although no significant changes were observed in HbA1c, insulin, and
TE

291 erythrocyte catalase activity [26].

292 We also hypothesized that the improvement in glycemic control after probiotic
EP

293 treatment could be mediated, in part, through immune-modulatory effects. To capture these
C

294 interrelations, were selected a set of key inflammatory and anti-inflammatory cytokines,
AC

295 including adipokines produced primarily by adipocytes (adiponectin), macrophages (resistin

296 and IL-6), or both (TNF-α), all related to glycemic control and insulin resistance [32-33].

297 Although other studies mention the relationship between inflammation and gut

298 microbiota [10, 34], few experimental and clinical studies investigated this association in the

299 diabetes context [5, 27, 35]. Some experimental studies reported reductions in inflammatory

300 cytokines, and/or improvements in glucose and insulin metabolims after ingestion of
13
ACCEPTED MANUSCRIPT
301 Lactobacillus spp during 14 wk [5, 35]. Two randomized trials did not observe effects on

302 inflammatory cytokines after the intake during 4 or 6 wk of L. acidophilus NCFM (1010

303 CFUs) or L. acidophilus, L. bulgaricus, L. bifidum, and L. casei (CFUs and strain not

304 reported), respectively [16, 27]. In agreement with these previous studies, IL-6 did not

305 change after intervention, however, TNF- α and resistin levels decreased in both treatments in

PT
306 our study. It has been shown that anti-inflammatory activities of goat milk oligosaccharides

RI
307 reside in their ability to function either as prebiotics or as a receptor for microorganisms

308 (Daddaoua et al., 2006). Others studies shows the ability of bioactive peptides derived from

SC
309 food fermentation in the immune system regulation, including inhibition of NF-κB, a pro-

310 inflammatory transcription factors [36-37].

311
U
In regard to anti-inflammatory cytokines (IL-10 and adiponectin), no changes were
AN
312 observed in the probiotic group, unlike the control group, which showed a significant
M

313 reduction at the end of trial. Thus, our results suggest that the reduction in these cytokines

314 reflected on lower glycemic control observed in the control group. Recently, Mohamadshahi
D

315 et al. [38], also reported that the consumption of probiotic yogurt enriched with L.
TE

316 acidophilus La-5 and B. lactis BB-12 (106 CFUs) for 8 wk caused significant decrease in

317 HbA1c and TNF-α levels in the intervention group, but no changes were observed in IL-6 and
EP

318 hs-CRP levels. In mice, L. rhamnosus GG orally administrated for 13 wk improves insulin
C

319 sensitivity by stimulating adiponectin secretion and consequent activation of AMPK [39],
AC

320 highlighting the important role of adiponectin in glycemic control. Some contradictory results

321 may be explained by complex regulation of cytokines, and we point out that the production of

322 IL-6 and IL-10 is stimulated by TNF-α, which was reduced in the present study.

323 Additionally, microbiota components account for the production of SCFA, which are

324 linked to anti-inflammatory mechanisms (inhibition of NF-κB) and also exerting a protective

325 function in favor of intestinal epithelium [40]. However, reports on the ability of lactic acid
14
ACCEPTED MANUSCRIPT
326 bacteria to modulate SCFA at the intestinal level are limited. In our literature review [41], we

327 didn’t find experimental or clinical trials that evaluated the effect of probiotics intake on the

328 SCFA in T2D context. Interestingly, growing evidence suggests a cross talk between SCFA

329 and improves in glycemic control, especially butyric and propionic acids via the G protein

330 coupled receptors FFA2 and FFA3 [42-43]. However, we found a significant increase in

PT
331 acetic acid after intervention in both groups, which has little effect on incretins (GLP-1, PYY,

RI
332 and GIP) secretion [42] compared to butyric and propionic acids. This increase in the acetic

333 acid in both treatments may be attributed to phenolic compounds present in FM [44].

SC
334 With regard to lipid profile, contradictory results have also been shown in the

335 literature. L. acidophilus La-5 and B. lactis BB-12 (106 CFUs) use contributed to the decrease

336
U
of TC (4.5%) and LDL-C (7.5%) levels (p < 0.05) after 6 wk in T2D subjects [45]. In a
AN
337 different study, the same probiotics taken for 8 wk, caused reductions in LDL-C/ HDL-C (p
M

338 = 0.01) and increase HDL-C levels [46]. However, other studies using different species of

339 Lactobacillus or B. coagulans and inulin for 6 wk did not observe effects on the lipid profile
D

340 in T2D subjects [16, 29]. Our study demonstrated that consumption of probiotic FM
TE

341 prevented a rise in TC and LDL-C compared to the control group, which can be attributed, in

342 part, by the higher total phenolic concentrations and antioxidant capacity in probiotic FM,
EP

343 however it was not enough to cause significant reductions in these parameters in the probiotic
C

344 group.
AC

345 Finally, probiotics as functional foods offer great potential to improve health and/or

346 help prevent certain diseases when taken as part of a balanced diet and healthy lifestyle. Our

347 results showed low intake of micronutrients and dietary fiber, and high intake of saturated

348 fatty acids (see Supplemental Table 4).

349 In conclusion, this trial suggests that probiotic consumption improves the glycemic

350 control in T2D subjects, however, the intake of fermented goat’s milk seems to be involved
15
ACCEPTED MANUSCRIPT
351 with changes in inflammatory cytokines (TNF-α and resistin) and in the acetic acid

352 concentrations. The major findings of this study are the following: (i) L. acidophilus and B.

353 lactis intake seem to be associated with a decrease of the fructosamine and HbA1c levels; (ii)

354 fermented goat’s milk consumption appears to be related to acetic acid content in faecal

355 samples; (iii) the role of probiotic in averting an increase of CT and LDL-C and a reduction

PT
356 of anti-inflammatory cytokines. Because our study was designed to be a short-term trial, our

RI
357 results need to be confirmed in long-term trials testing the hypothesis that probiotic

358 supplementation is effective in improving glycemic control, regulating SCFA levels and

SC
359 decreasing oxidative stress in T2D subjects.

360 Conflict of interest

361 None.
U
AN
362 Acknowledgments
M

363 This study was supported by Brazilian Agricultural Research Corporation

364 (EMBRAPA) and Foundation for Research Support of the State of Minas Gerais
D

365 (FAPEMIG). We thank study volunteers for their participation; Mariana Rodrigues and
TE

366 Etianne Lopes (INTA) for trial staff; Adriano Lima (EMBRAPA) for providing data

367 management support; Renata Toledo (Federal University of Viçosa) and José Ricardo
EP

368 (EMBRAPA) for providing laboratory support. We thank Tabosa Santos and Isabel Cristina
C

369 (EMBRAPA) for technical assistance and Marcelo Amadei for providing study recruitment
AC

370 support. The authors also thank the Embrapa Goats and Sheep for generous support.

371 Authors contributions

372 LBT, KMOS, and HSDM designed the trial. LBT and KMOS were responsible for the

373 analysis and production of fermented milk. LBT and HSDM conducted the reseach- were

374 responsible for study recruitment, screening, and delivery of interventions. LBT and HSDM

375 did the statistical analysis. LBT and LLO analysed and interpreted stress oxidative datas. All
16
ACCEPTED MANUSCRIPT
376 authors participated in data interpretation. LBT wrote the first draft of the report, and all other

377 authors commented on the draft and approved the final version. Authorship order reflects

378 overall contribution to the work presented. None of the authors had a conflict of interest.

379 Appendix A. Supplementary data

PT
RI
U SC
AN
M
D
TE
C EP
AC
17
ACCEPTED MANUSCRIPT
References

[1] IDF. IDF Diabetes Atlas. In: 6, editor. International Diabetes Federation. 6 ed2013.

[2] Kershaw EE, Flier JS. Adipose Tissue as an Endocrine Organ. The Journal of Clinical

Endocrinology & Metabolism. 2004;89:2548-56.

[3] Rani AJ, Mythili SV. Study on Total Antioxidant Status in Relation to Oxidative Stress in

PT
Type 2 Diabetes Mellitus. J Clin Diagn Res. 2014;8:108-10.

RI
[4] Muoio DM, Newgard CB. Molecular and metabolic mechanisms of insulin resistance and

[beta]-cell failure in type 2 diabetes. Nat Rev Mol Cell Biol. 2008;9:193-205.

SC
[5] Hsieh F-C, Lee C-L, Chai C-Y, Chen W-T, Lu Y-C, Wu C-S. Oral administration of

Lactobacillus reuteri GMNL-263 improves insulin resistance and ameliorates hepatic

U
steatosis in high fructose-fed rats. Nutrition & Metabolism. 2013;10:35.
AN
[6] Wang S, Zhu H, Lu C, Kang Z, Luo Y, Feng L, et al. Fermented milk supplemented with
M

probiotics and prebiotics can effectively alter the intestinal microbiota and immunity of host

animals. Journal of Dairy Science. 2012;95:4813-22.


D

[7] Larsen N, Vogensen FK, van den Berg FWJ, Nielsen DS, Andreasen AS, Pedersen BK, et
TE

al. Gut Microbiota in Human Adults with Type 2 Diabetes Differs from Non-Diabetic Adults.

PLoS ONE. 2010;5:e9085.


EP

[8] Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of


C

gut microbiota in type 2 diabetes. Nature. 2012;490:55-60.


AC

[9] Karlsson FH, Tremaroli V, Nookaew I, Bergstrom G, Behre CJ, Fagerberg B, et al. Gut

metagenome in European women with normal, impaired and diabetic glucose control. Nature.

2013;498:99-103.

[10] Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic

endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761-72.


18
ACCEPTED MANUSCRIPT
[11] Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, et al. Gut

microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide

production with consequences for appetite sensation and glucose response after a meal. The

American Journal of Clinical Nutrition. 2009;90:1236-43.

[12] Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, et al. The

PT
short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid

RI
receptor 2 in rodents. Int J Obes. 2014.

[13] Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of the

SC
intestinal microbiota and immune system. Nature. 2012;489:231-41.

[14] Cani P, Neyrinck A, Fava F, Knauf C, Burcelin R, Tuohy K, et al. Selective increases of

U
bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a
AN
mechanism associated with endotoxaemia. Diabetologia. 2007;50:2374 - 83.
M

[15] ADA. American Diabetes Association. Standards of Medical Care in Diabetes—2014.

Diabetes Care. 2014;37:S14-S80.


D

[16] Mazloom Z, Yousefinejad A, Dabbaghmanesh MH. Effect of probiotics on lipid profile,


TE

glycemic control, insulin action, oxidative stress, and inflammatory markers in patients with

type 2 diabetes: a clinical trial. Iranian journal of medical sciences. 2013;38:38-43.


EP

[17] Buriti FCA, Freitas SC, Egito AS, dos Santos KMO. Effects of tropical fruit pulps and
C

partially hydrolysed galactomannan from Caesalpinia pulcherrima seeds on the dietary fibre
AC

content, probiotic viability, texture and sensory features of goat dairy beverages. LWT - Food

Science and Technology. 2014;59:196-203.

[18] Peryam DR, Pilgrim FJ. Hedonic scale method of measuring food preferences. Food

Technology. 1957;11, Suppl.:9-14.


19
ACCEPTED MANUSCRIPT
[19] Geloneze B, Repetto EM, Geloneze SR, Tambascia MA, Ermetice MN. The threshold

value for insulin resistance (HOMA-IR) in an admixtured population IR in the Brazilian

Metabolic Syndrome Study. Diabetes Research and Clinical Practice 2006;72:219-20.

[20] Vignali DAA. Multiplexed particle-based flow cytometric assays. Journal of

Immunological Methods. 2000;243:243-55.

PT
[21] Smiricky-Tjardes MR, Grieshop CM, Flickinger EA, Bauer LL, Fahey GC. Dietary

RI
galactooligosaccharides affect ileal and total-tract nutrient digestibility, ileal and fecal

bacterial concentrations, and ileal fermentative characteristics of growing pigs. Journal of

SC
Animal Science. 2003;81:2535-45.

[22] Gravitz L. Microbiome: The critters within. Nature. 2012;485:12-3.

U
[23] Tabuchi M, Ozaki M, Tamura A, Yamada N, Ishida T, Hosoda M, et al. Antidiabetic
AN
effect of lactobacillus GG in streptozotocin-induced diabetic rats. Biosci Biotechnol
M

Biochem. 2003;67:1421 - 4.

[24] Yadav H, Jain S, Sinha P. Antidiabetic effect of probiotic dahi containing lactobacillus
D

acidophilus ans lactobacillus casei in high fructose fed rats. Nutr. 2007;23:62 - 8.
TE

[25] Kang J-H, Yun S-I, Park M-H, Park J-H, Jeong S-Y, Park H-O. Anti-Obesity Effect of

Lactobacillus gasseri BNR17 in High-Sucrose Diet-Induced Obese Mice. PLoS ONE.


EP

2013;8:e54617.
C

[26] Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, Niafar M, Asghari-Jafarabadi M, Mofid


AC

V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition

(Burbank, Los Angeles County, Calif). 2012;28:539-43.

[27] Andreasen AS, Larsen N, Pedersen-Skovsgaard T, Berg RMG, Møller K, Svendsen KD,

et al. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic

inflammatory response in human subjects. British Journal of Nutrition. 2010;104:1831-8.


20
ACCEPTED MANUSCRIPT
[28] Asemi Z, Zare Z, Shakeri H, Sabihi S, Esmaillzadeh A. Effect of Multispecies Probiotic

Supplements on Metabolic Profiles, hs-CRP, and Oxidative Stress in Patients with Type 2

Diabetes. Annals of Nutrition and Metabolism. 2013;63:1-9.

[29] Asemi Z, Khorrami-Rad A, Alizadeh S-A, Shakeri H, Esmaillzadeh A. Effects of

synbiotic food consumption on metabolic status of diabetic patients: A double-blind

PT
randomized cross-over controlled clinical trial. Clinical Nutrition. 2014;33:198-203.

RI
[30] Stephens JW, Khanolkar MP, Bain SC. The biological relevance and measurement of

plasma markers of oxidative stress in diabetes and cardiovascular disease. Atherosclerosis.

SC
2009;202:321-9.

[31] Wang X, Bao W, Liu J, OuYang Y-Y, Wang D, Rong S, et al. Inflammatory Markers

U
and Risk of Type 2 Diabetes: A systematic review and meta-analysis. Diabetes Care.
AN
2013;36:166-75.
M

[32] Tuttolomondo A, La Placa S, Di Raimondo D, Bellia C, Caruso A, Lo Sasso B, et al.

Adiponectin, resistin and IL-6 plasma levels in subjects with diabetic foot and possible
D

correlations with clinical variables and cardiovascular co-morbidity. Cardiovascular


TE

Diabetology. 2010;9:50.

[33] Fasshauer M, Paschke R. Regulation of adipocytokines and insulin resistance.


EP

Diabetologia. 2003;46:1594-603.
C

[34] Magrone T, Jirillo E. The interaction between gut microbiota and age-related changes in
AC

immune function and inflammation. Immunity & Ageing. 2013;10:31.

[35] Matsuzaki T, Yamazaki R, Hashimoto S, Yokokura T. Antidiabetic effects of an oral

administration of lactobacillus casei in a non-insulin-dependent diabetes mellitus (NIDDM)

model using KK-Ay mice. Endocr J. 1997;44:357 - 65.


21
ACCEPTED MANUSCRIPT
[36] Martínez-Augustin O, Rivero-Gutiérrez B, Mascaraque C, Sánchez de Medina F. Food

Derived Bioactive Peptides and Intestinal Barrier Function. International Journal of

Molecular Sciences. 2014;15:22857-73.

[37] Marcone S, Haughton K, Simpson P, Belton O, Fitzgerald D. Milk-derived bioactive

peptides inhibit human endothelial-monocyte interactions via PPAR-gamma dependent

PT
regulation of NF-kappaB. Journal of Inflammation. 2015;12:1.

RI
[38] 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

SC
diabetes. Bioimpacts. 2014;4:83-8.

[39] Kim S-W, Park K-Y, Kim B, Kim E, Hyun C-K. Lactobacillus rhamnosus GG improves

U
insulin sensitivity and reduces adiposity in high-fat diet-fed mice through enhancement of
AN
adiponectin production. Biochemical and Biophysical Research Communications.
M

2013;431:258-63.

[40] De Vuyst L, Leroy F. Cross-feeding between bifidobacteria and butyrate-producing


D

colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas


TE

production. International Journal of Food Microbiology. 2011;149:73-80.

[41] Tonucci LB, Santos KMO, Ferreira CLLF, Ribeiro SMR, Oliveira LL, Martino HSD.
EP

Gut Microbiota and Probiotics: Focus on Diabetes Mellitus. Critical Reviews in Food Science
C

and Nutrition. 2015.


AC

[42] Lin HV, Frassetto A, Kowalik Jr EJ, Nawrocki AR, Lu MM, Kosinski JR, et al. Butyrate

and Propionate Protect against Diet-Induced Obesity and Regulate Gut Hormones via Free

Fatty Acid Receptor 3-Independent Mechanisms. PLoS ONE. 2012;7:e35240.

[43] Kasubuchi M, Hasegawa S, Hiramatsu T, Ichimura A, Kimura I. Dietary Gut Microbial

Metabolites, Short-chain Fatty Acids, and Host Metabolic Regulation. Nutrients.

2015;7:2839-49.
22
ACCEPTED MANUSCRIPT
[44] Parkar SG, Trower TM, Stevenson DE. Fecal microbial metabolism of polyphenols and

its effects on human gut microbiota. Anaerobe. 2013;23:12-9.

[45] Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, Niafar M, Asghari-Jafarabadi M, Mofid

V, et al. Effect of probiotic yogurt containing Lactobacillus acidophilus and Bifidobacterium

lactis on lipid profile in individuals with type 2 diabetes mellitus. Journal of dairy science.

PT
2011;94:3288-94.

RI
[46] Mohamadshahi M, Veissi M, Haidari F, Javid AZ, Mohammadi F, Shirbeigi E. Effects

of probiotic yogurt consumption on lipid profile in type 2 diabetic patients: A randomized

SC
controlled clinical trial. J Res Med Sci. 2014;19:531-6.

U
AN
M
D
TE
C EP
AC
23
ACCEPTED MANUSCRIPT
Tables

Table 1. Baseline characteristics of the type 2 diabetes participants

Characteristics Control Probiotic p1 value

(n = 22) (n = 23)

PT
Age (y) 50.95 ± 7.20 51.83 ± 6.64 0.73

Sex (M/F) 14/8 (63/37) 12/11 (53/47) 0.45

RI
Diabetes duration (y) 4.5 (2 – 15) 6.0 (2 -17) 0.51

SC
Regular physical activity 12 (54.5) 09 (39.1) 0.31

U
Weight (kg) 77.15 ± 13.85 71.70 ± 12.43 0.16
AN
BMI (kg/m2) 27.94 ± 4.15 27.49 ± 3.97 0.65

Total body fat (%) 32.95 ± 8.96 33.92 ± 8.24 0.70


M

HbA1c (%) 5.35 (4.86 – 6.15) 6.07 (5.39 – 7.0) 0.04*


D

FPG (mmol/L) 7.38 ± 2.39 7.97 ± 2.31 0.41


TE

HOMA-IR 2.15 (1.71 – 3.26) 2.63 (1.57 – 3.51) 0.55


EP

Metformin (mg) 997.74 ± 460.50 1123.91 ± 578.76 0.36

Glibenclamide (mg) 5.02 ± 12.60 9.78 ± 23.84 0.83


C
AC

Data are mean ± SD, n (%) or median (P25-P75). 1Significant difference between

groups at baseline (p < 0.05 from Student’s t test or Mann–Whitney test). M =

masculine; F = female; BMI = body mass index; HbA1c = haemoglobin A1c; FPG =

fasting plasma glucose.


ACCEPTED MANUSCRIPT

24

Table 2: Metabolic parameters of type 2 diabetes subjects at baseline and endpoint by fermented milk treatment

Control (n = 22) Probiotic (n = 23) Intervention effect

PT
Measure Mean/media
Week 0 Week 6 Mean/median change p1 Week 0 Week 6 p1 Effect (95% CI) p2
n change

RI
FPG, mmol/L 7.38 (2.37) 7.54 (2.50) 0.16 0.65 7.89 (2.28) 8.41 (2.41) 0.52 0.14 6.35 (- 0.64 to 1.34) 0.48

SC
Fru, mmol/L 295.5 (62.23) 296.90 (59.64) 1.36 0.85 302.82 (51.31) 292.90 (53.25) -9.91 0.04 -12.06 (-28.51 to 4.40) 0.15

U
5.66 (4.9 to ..
HbA1c, % 5.35 (4.9 to 6.1) 0.11 0.82 6.07 (5.4 to 7.0) 5.40 (5.1to 7.3) -0.67 0.06 0.02

AN
6.7)

6.65 (5.2 to ..

M
Insulin, µU/mL 8.3 (5.0 to 10.1) -1.65 0.95 7.5 (5.2 to 11.9) 6.8 (5.2 to 11.4) -0.70 0.73 0.72
11.4)

D
2.15 (1.71 to 2.3 (1.55 to 2.63 (1.57 to 2.65 (1.71 to ..

TE
HOMA-IR 0.15 0.73 0.02 0.41 0.77
3.26) 3.27) 3.52) 4.21)
EP
TC, mmol/L 4.85 (1.32) 5.30 (1.19) 0.55 0.01 4.66 (1.38) 4.51 (1.11) - 0.15 0.52 -0.58 (-1.13 to -0.003) 0.04

LDL-C, mmol/L 2.24 (1.24) 2.60 (1.11) 0.36 0.04 2.31 (1.17) 2.11 (0.82) - 0.20 0.31 -0.52 (-1.02 to -0.03) 0.03
C
AC

HDL-C, -0.006 (-0.21 to 0.08)


1.51 (0.26) 1.53 (0.34) 0.02 0.59 1.56 (0.29) 1.53 (0.33) - 0.03 0.50 0.38
mmol/L

TC:HDL-C 3.20 (1.06) 3.48 (0.97) 0.28 0.03 2.98 (0.64) 2.94 (0.72) - 0.05 0.70 -0.17 (-0.53 to 0.19) 0.35
ACCEPTED MANUSCRIPT

25

LDL-C:HDL-C 1.48 (0.93) 1.69 (0.83) 0.21 0.08 1.48 (0.64) 1.37 (0.58) - 0.11 0.30 -0.22 (-0.56 to 0.12) 0.19

TAG, mmol/L 1.83 (0.72) 1.99 (0.91) 0.16 0.73 1.60 (0.63) 1.68 (0.63) 0.08 0.28 -0.008 (-0.44 to 0.27) 0.62

TAS, (mM) 0.31 (0.12) 0.33 (0.08) 0.02 0.23 0.31 (0.09) 0.33 (0.11) 0.02 0.39 0.01 (-0.07 to 0.08) 0.87

PT
F2-iso, (pg/mL) 62.99 (33.67) 63.42 (32.35) - 0.43 0.94 74.66 (30.20) 72.49 (34.70) - 2.17 0.76 -2.59 (-21.41 to 16.23) 0.78

RI
Data are means (SD) or median (P25-P75). 1Difference between baseline and endpoint. p value obtained from paired t test/Wilcoxon matched-pairs signed-rank test for the

SC
within-group comparisons. 2Obtained from unpaired Student’s t test or Mann–Whitney test, as statistical differencece between changes (control vs probiotic). FPG = fasting

blood glucose; Fru = fructosamine; TC = total cholesterol; TAG = triglyceride; TAS = total antixodant status; F2-iso = F2-isoprostane.

U
AN
M
D
TE
C EP
AC
26
ACCEPTED MANUSCRIPT
Figures

PT
RI
U SC
AN
M
D
TE
EP

Fig 1. Trial profile.


C
AC
27
ACCEPTED MANUSCRIPT

PT

RI
U SC
AN
*
*
† *
M
D
TE
C EP
AC

Fig 2. Effects of the fermented milks intake on cytokine levels

Data are shown as mean (SD). *p < 0.05, †p = 0.001 from paired t test or for Wilcoxon

matched-pairs signed-rank test, as statistical within group differences (baseline vs endpoint).

IL = interleukin; TNF- α = tumor necrosisfactor alpha.


28
ACCEPTED MANUSCRIPT

PT
RI
SC
Fig 3. Effects of the fermented milks intake on faecal short-chain fatty acid

U
concentrations

Data are shown as mean (SD). †p ≤ 0.01 from Wilcoxon matched-pairs signed-rank test, as
AN
statistical within group differences (baseline vs endpoint).
M
D
TE
C EP
AC

Vous aimerez peut-être aussi