You are on page 1of 9

Received: 5 July 2016 | Revised: 12 August 2016 | Accepted: 17 August 2016

DOI 10.1111/jfbc.12336

FULL ARTICLE

b-Glucan, but not Lactobacillus plantarum P-8, inhibits lipid


accumulation through selected lipid metabolic enzymes in
obese rats

Hsiu-Chuan Lee1 | Wei-Ting Yu1 | Yu-Ru Guo1 | Shih-Yi Huang1,2

1
School of Nutrition and Health Sciences,
Taipei Medical University, Taipei, Taiwan
Abstract
2
Graduate Institute of Metabolism and We compared the effects of barley b-glucan and Lactobacillus plantarum P-8 on changes in body
Obesity Science, Taipei Medical University, weight, white adipose tissue, and the activities of specific lipid metabolic enzymes in diet-induced
Taipei, Taiwan obese rats. Obese rats were administered barley b-glucan (B), L. plantarum P-8 (P), or both for
Correspondence 8 weeks. The results showed that the dietary intervention reduced the body weight and white adi-
Shih-Yi Huang, School of Nutrition and
pose tissue accumulation, and altered gut microbiota in the distal faeces of the B group.
Health Sciences, Taipei Medical University,
250 Wu-Xing Street, Taipei 110, Taiwan. Furthermore, acetate and propionate levels considerably increased in the caecal digest and rectal
Email: Sihuang@tmu.edu.tw feces of the B group. In addition, b-glucan, but not L. plantarum P-8, increased the activities of
Funding information hepatic acetyl CoA carboxylase, fatty acid synthase, and carnitine palmitoyl transferase and
Ministry of Science and Technology, reduced epididymal fat lipoprotein lipase activity, thus resulting in decreased white adipose tissue
Taiwan, Grant/Award Number: NSC101-
accumulation in the obese rats. However, b-glucan and L. plantarum P-8 showed no synergistic
2320-B-038-020-MY2; NSC102-2313-
B-038-003-MY2, and MOST103-2320- effects.
B-038-012-MY3
Practical applications
b-Glucan ameliorates hyperlipidaemia, increases hepatic acetyl CoA carboxylase, fatty acid syn-
thase, and carnitine palmitoyl transferase activities, and reduces epididymal fat lipoprotein lipase
activity, thus resulting in reduced white adipose tissue accumulation in obese rats. b-Glucan
reduces of reducing lipid levels and regulating specific lipid metabolic enzymes.

KEYWORDS
beta-glucan, Lactobacillus plantarum, obesity, short chain fatty acids

1 | INTRODUCTION Kaser, 2009) and are involved in diseases such as obesity and meta-
bolic disorders (Tilg & Moschen, 2014). Gut microbiota ferment indi-
Incidences of overweight and obesity have been increasing, and have gestible polysaccharides into short-chain fatty acids (SCFAs), which are
thus become a major health concern worldwide (WHO, 2016). The then partially absorbed into the colon and mainly excreted in faeces
World Health Organization indicated that at least 2.3 billion adults (Wong, de Souza, Kendall, Emam, & Jenkins, 2006). An increase
were overweight, and more than 700 million were obese, in 2015. observed in the faecal SCFA levels of germ-free mice colonised with
Obesity increases the risk of several metabolic disorders, including the microbiota of obese mice indicates that gut microbiota-produced
complex dyslipidaemia, hypertension, atherosclerosis, glucose intoler- SCFAs may have a critical role in obesity development (Turnbaugh
ance, and type 2 diabetes mellitus, as well as the risk of mortality et al., 2006). Decreased levels of gut microbiota such as Methanobrevi-
(Ogden, Carroll, & Flegal, 2014). Thus, obesity prevention is being con- bacter smithii and reduction in the Bacteroidetes/Firmicutes ratio have
sidered in public health strategies and during functional food been strongly associated with obesity in humans (Ley et al., 2005;
development. Turnbaugh et al., 2009).
Recent studies have suggested that gut microbiota and dietary Dietary fibre consumption has been demonstrated to produce
fibre are critical for harvesting energy from the diet (Tilg, Moschen, & beneficial health effects and improvements in healthy participants and

J Food Biochem. 2017;41:e12336. wileyonlinelibrary.com/journal/jfbc V


C 2016 Wiley Periodicals, Inc. | 1 of 9
https://doi.org/10.1111/jfbc.12336
2 of 9 | LEE ET AL.

patients with metabolic syndrome (Carlson, Eisenmann, Norman, Ortiz, TA BL E 1 Compositions of experimental diets
& Young, 2011; Hu, 2003). Oat or barley b-glucans may reduce total
Groups Normal diet High-fat diet
cholesterol (TC) levels in participants with hypercholesterolemia (FDA,
Energy (%)
1997, 21 January). Gut microbiota-produced SCFAs from dietary fibre
Protein 19 19
may affect energy homeostasis, neurotransmission, and even disease Fat 10.2 44.8
development (Wu, Tremaroli, & Backhed, 2015). b-glucan has dual Carbohydrate 70.8 36.2
kcal/g 3.6 4.6
health benefits, accelerating bile acid excretion (Ellegard & Andersson,
2007) and functioning as a prebiotic to produce SCFA and regulate Subgroups N C P B M

energy homeostasis (Wong et al., 2006). Several interventional studies Ingredients (g/Kg)
Casein 177 224 223 223 223
on probiotics have reported that gut microbiota-produced SCFAs affect
L-cystine 3 3 2 2 2
lipid metabolism and reduce body weight in patients with obesity Cornstarch 541 187 187 151 137
(Druart et al., 2014; Salazar et al., 2015). Maltodextrin 30 30 30 30 30
Sucrose 100 215 214 214 214
Studies have suggested that SCFA production by gut microbiota is Cellulose 50 50 50 50 50
associated with dietary fibre consumption, and that these SCFAs may be Soybean oil 22 22 21 21 21
Lard 22 216 214 214 214
involved in the initiation of satiety, energy homeostasis, inflammation,
AIN 93M mineral mix 35 35 35 35 35
and chronic metabolic dysfunction (Byrne, Chambers, Morrison, & Frost, Calcium phosphate, dibasic 2 2 2 2 2
2015b; Druart et al., 2014). Most of the relevant studies have focused AIN 93 vitamin mix 15 15 15 15 15
Choline bitartrate 3 3 3 3 3
on the prebiotic potential of indigestible fructo-oligosaccharides (i.e., inu-
b-Glucan 0 0 0 40 40
lin) and galacto-oligosaccharides for increasing the total quantity of gut L. plantarum 0 0 4 0 4
microbiota. The role of mixed-linkage b-glucans fermented by gut micro- N 5 normal; C 5 control; P 5 Lactobacillus plantarum; B 5 (1,3/1,4)
biota in increasing SCFA levels has gained little attention (Zhong, Marun- b-glucan; M 5 Lactobacillus plantarum plus b-glucan.
gruang, Fak, & Nyman, 2015). Thus, with respect to the food matrix,
b-glucan-containing foods may contribute to optimal growth conditions
for various gut microbial strains and increase SCFA production (Arena fat diet supplemented with barley b-glucan at 1 g/d (B), L. plantarum P-8
et al., 2014; Nielsen et al., 2016). (1 3 109 colony-forming units [CFU]) at 0.1 g/d (P), L. plantarum P-8 at
Beneficial bacteria, such as the members of the Lactobacillus and 0.1 g/d plus b-glucan at 1 g/d (M), or no supplements, as a control (C),
Bifidobacterium genera, may have anti-obesity (Park, Oh, & Cha, 2014; for 8 weeks. The composition of the treatment diets was based on the
Yoo et al., 2013) and anti-inflammatory (Zhong et al., 2015) properties. American Institute of Nutrition-93M diet (MP Biomedicals, LLC Santa

However, few studies have evaluated the synergistic effects of prebiot- Ana, CA, USA), containing 10% and 45% soybean oil (Table 1). Barley

ics and probiotics on metabolic dysfunction diseases and the status of (1!3,1!4)-b-glucan (GlucagelTM; 75%; average molecular weight, 740

microbiota-produced SCFAs in the gut. Therefore, the aim of this study kDa) was purchased from DKSH Taiwan Ltd, Taiwan. L. plantarum P-8

is to evaluate the effects of barley b-glucan and Lactobacillus plantarum (LABCOTTM lactic acid bacteria GFB007) was purchased from GeneFerm

P-8 on gut microbiota, SCFA production, body fat distribution, and the Biotechnology Co., Ltd, Tainan, Taiwan. During the experiment, the food

activities of specific lipid metabolic enzymes in diet-induced obese rats. intake and body weight were recorded and then used to determine the
food efficiency of diet-induced obesity. All animals were provided the
aforementioned diets and water ad libitum. These conditions were main-
2 | MATERIALS AND METHODS
tained throughout all the experiments. All animal procedures complied
with the guidelines of the Institutional Animal Care and Use Committee
2.1 | Animal protocols and experimental design
of Taipei Medical University (LAC-101-0190).
Fifty 6-week-old male Sprague Dawley rats (BioLASCO Taiwan, Taipei, Fresh rectal faeces from the obese rats were collected after mas-
Taiwan), each weighing 150–180 g, were housed in a temperature- and saging their stomachs at the beginning of week 0 and the end of week
humidity-controlled room with a 12-h light–dark cycle (light period, 8 of the intervention. The fresh rectal faeces (approximately 10 g) col-
08:00–20:00; temperature 228C–248C; humidity, 60%; two rats per lected from the treated mice were frozen in liquid nitrogen and then
cage). The rats were allowed to adapt for 2 weeks and fed Rodent Lab- stored in a 2808C freezer for gut microbiota and SCFAs analyses.
oratory Chow 5001. At the end of the experiment, all the rats were euthanised using
The rats were then assigned to the following diet groups for 12 carbon dioxide, and their livers, kidneys, and caeca were excised and
weeks: a normal diet group with fat constituting 10% of total energy weighed prior to further biochemical analysis. Furthermore, adipose tis-
(n 5 10) and a high-fat diet group intended to induce obesity with fat sues, including inguinal, retroperitoneal, epididymal, perirenal, and mes-
constituting 45% of total energy (n 5 40) (Ghibaudi, Cook, Farley, van enteric fat were collected and weighed prior to further analysis.
Heek, & Hwa, 2002). The rats fed the high-fat diet were considered Abdominal blood was collected for biochemical analyses. Caecal epi-
obese when their body weight was >10% of that of the rats fed the nor- thelial tissue, colonic mucin, and rectal faeces were collected to analyse
mal diet (Abdul Kadir, Rahmat, & Jaafar, 2015). The 40 obese rats were SCFA concentrations. All chemicals used in this study were obtained
then divided into four groups each of which was provided either a high- from Sigma-Aldrich (St. Louis, MO, USA).
LEE ET AL. | 3 of 9

2.2 | Biochemical assays tration was determined using a DC protein assay kit II (Bio-Rad Labora-
tories, Berkeley, CA, USA); bovine serum albumin (Pierce, Rockford, IL,
To evaluate the safety of the experimental diets, overnight tail-vein
USA) was used to generate a standard curve. The ELISA microplates
blood samples were collected in ethylenediaminetetraacetic acid
were read using an ELISA reader (Dynatech Laboratories, Chantilly, VA,
(EDTA) tubes, centrifuged at 3,000 g for 10 min to obtain plasma, and
USA), with a maximum absorbance of 450 nm.
maintained at 2808C at the end of the experiment. The activities of
The lipoprotein lipase (LPL) activity of epididymal fat in the adipose
glutamate oxalate transferase (GOT) and glutamate pyruvate transfer-
tissue specimens was evaluated by using a fluorescent method, accord-
ase (GPT) and levels of blood urea nitrogen, creatinine, triglycerides
ing to the manufacturer’s instructions (LPL activity Kit, Roar Biomedi-
(TGs), TC, low-density lipoprotein cholesterol (LDLc), and high-density
cal, New York, NY). A total of 0.2 g of tissues was mixed and
lipoprotein cholesterol (HDLc) were measured from the plasma sam-
homogenised with 2 mL of a homogenising buffer (containing 0.3 M
ples. GOT and GPT activities and blood urea nitrogen and creatinine
mannitol, 10 mM HEPES, 1 mM EGTA, pH 7.2) in an ice bath. The
levels were analysed by using an automatic chemistry analyser (Hitachi
homogenates were then centrifuged at 1,200 g and 48C for 10 min,
7170S). TG and TC levels were measured with an Ortho Clinical Diag-
and the supernatant was centrifuged at 13,000 g and 48C for 12 min.
nostics VITROS® 950 automated analyser (Johnson & Johnson, New
The pellets were then resuspended in 0.07 M sucrose, 0.22 M manni-
Brunswick, NJ, USA), and LDLc and HDLc levels were determined by
tol, 0.02 M HEPES buffer (pH 7.4), and 0.01 M EDTA. The superna-
using a TBA-c16000 automated analyser (Toshiba, Tokyo, Japan). A
tants were used for LPL activity assay (Notarnicola, Miccolis, Tutino,
modified version of Kumar’s experimental method (Kumar, Grover, &
Lorusso, & Caruso, 2012). An aliquot of the supernatant (100 lL) was
Batish, 2011) was used to extract faecal cholic acid, the concentration
incubated with 100 mL of prediluted substrate emulsion at 378C for
of which was measured using a bile acid kit (BXC0581A, Fortress Diag-
1 h. The hydrolysed TGs were measured at 370-nm excitation and
nostics, Antrim, UK). Faecal water content was calculated using the fol-
450-nm emission. The fluorescence intensity values of the samples
lowing equation: water content (%) 5 (wet weight2dry weight)/wet
were compared with the values of a standard curve in each run. LPL
weight 3 100.
activity was expressed as hydrolysed substrate nmol/min/mg protein,
and evaluated using the Lowry method. All measurements were per-
2.3 | Selected lipid metabolic enzyme activities formed in duplicate.
The activities of hepatic lipid-regulating enzymes, namely acetyl CoA
carboxylase (ACC), fatty acid synthase (FAS), and carnitine palmitoyl 2.4 | Faecal sample culture
transferase (CPT)21, were assayed using a previously described spec-
The faeces were collected from the obese rats and frozen at weeks
trophotometric method (Numa, Bortz, & Lynen, 1965; Small, Burdett, &
0 and 8, on the same day as the gut microbiota and SCFAs analyses. In
Connock, 1985). Five hundred mg fresh liver samples were mixed and
the gut microbiota analysis, 1 g of the faecal sample was dissolved in
homogenised with 5 mL of a homogenising buffer (containing 0.3 M
9 mL of saline and homogenised. Subsequently, the faecal water was vor-
mannitol, 10 mM HEPES, 1 mM EDTA, pH 7.2) at 48C. The hepatic
texed for 10 min and dilutions were prepared. The faecal water samples
homogenates were centrifuged at 1,200 g and 48C for 10 min, and the
were then incubated in duplicate at various concentrations. The faecal
supernatant was again centrifuged at 7,200 g and 48C for 12 min. Pel-
samples were cultured on media specific for Escherichia coli (Endo agar),
lets were then resuspended in 0.07 M sucrose, 0.22 M mannitol,
Lactobacillus (MRS agar), Bifidobacterium (BIM agar), and total anaerobic
0.02 M HEPES buffer (pH 7.4), and 0.01 M EDTA. The resulting post-
bacteria (CDC agar) for 48 h at 378C under anaerobic conditions. Follow-
nuclear supernatants were assayed for ACC, FAS, and CPT-1 activities.
ing the incubation, the various groups of bacteria on the media were
FAS activity was measured as malonyl CoA-dependant oxidation by
counted. The results are expressed as CFU per gramme of wet faeces.
following the decrease in the absorbance at 340 nm resulting from
NADPH oxidation in the presence of acetyl-CoA. The FAS activity was
2.5 | SCFA analysis
expressed as the nmol of NADPH that was reduced per min per mg
protein (nmol/min/mg protein). ACC activity was determined as The caecal epithelial tissue and faecal samples were collected to ana-
described previously (Numa et al., 1965). CPT-1 activity was deter- lyse SCFA concentrations. SCFA extraction procedures were modified
mined on the basis of the method developed by Small et al. (1985) as described previously (Ezz El-Arab, 2009). All faecal samples for each
with minor modifications. The tissue homogenates were centrifuged at group and time interval were ground and thoroughly mixed for SCFA
800 g and 48C for 10 min, and the supernatant was centrifuged at analysis. A portion of the faecal sample (100 mg) was mixed with 500
7,200 g at 48C for 12 min. The pellets were then resuspended in mL of deionised water and 4-methyl-n-valeric acid (as an internal stand-
0.07 M sucrose, 0.22 M mannitol, 2 mmol/L HEPES buffer (pH 7.4), ard), added to a 2-mL Eppendorf tube, and mixed for 2 min. The mixed
and 1 mmol/L EDTA. The assay was based on the moiety CoA of pal- sample was centrifuged at 10,000 rpm and 48C for 1 min. The superna-
mitoyl CoA released by CPT-1 and reacted with 5,50 -dithiobis-(2-nitro- tant was transferred to another 2-mL Eppendorf tube. Subsequently,
benzoic acid) at 378C. The changes in the absorbance at 412 nm were 20 mL of 20% trichloroacetic acid solution was mixed with the superna-
measured at 20-s intervals for 5 min. The activity was expressed as tant, and the mixture was centrifuged at 10,000 rpm at 48C for 25 min.
CoASH nmol/min/mg protein (Small et al., 1985). The protein concen- SCFAs were extracted from the supernatant two times by using 1 mL
4 of 9 | LEE ET AL.

TA BL E 2 Dietary intake and weight of selected adipose tissues in each group

N C P B M

Dietary intake (g) 20.8 6 0.8 20.8 6 1.2 21.0 6 0.7 20.0 6 1.1 20.9 6 0.5

Body weight gain (g) 68.9 6 10.0 124.6 6 10.6* a


130.1 6 11.1 a
76.1 6 5.3 b
113.3 6 9.9a
Liver weight (g) 13.5 6 0.7 14.5 6 0.9 16.9 6 1.3 14.2 6 0.8 12.8 6 0.7

Total body fat (g) 45.8 6 3.5 98.3 6 6.6*ab 112.8 6 10.8a 87.6 6 5.9b 101.4 6 7.6ab
% of body weight 8.0 6 0.4 13.3 6 0.6*ab 14.5 6 1.1a 12.5 6 0.5b 13.7 6 0.7ab
Retroperitoneal fat (g) 3.11 6 0.53 3.11 6 0.24 2.45 6 0.17 2.28 6 0.19 3.41 6 0.41
Epididymal fat (g) 9.8 6 1.0 20.8 6 1.5* 23.0 6 2.0 19.5 6 1.7 21.1 6 1.8
Perirenal fat (g) 17.5 6 1.4 38.7 6 2.7*a 49.0 6 5.8b 35.4 6 3.5a 42.9 6 3.5a
Inguinal fat (g) 5.2 6 0.4 12.3 6 1.3*a 16.1 6 1.5b 11.5 6 0.7a 11.2 6 1.1a
Mesenteric fat (g) 10.3 6 0.9 23.5 6 2.4*a 22.9 6 2.0ab 18.9 6 1.6b 22.3 6 1.9ab

Note. Data are expressed as the mean 6 SEM, n 5 8–10.


N 5 normal; C 5 control; P 5 Lactobacillus plantarum; B 5 (1,3/1,4) b-glucan; M 5 Lactobacillus plantarum plus b-glucan.
The superscript symbol “*” means significantly different from the normal group (p < .05). Values in the same row with different superscript letters signif-
icantly differ at p < .05 by one-way ANOVA with least significant difference.

of ether. The ether layer was collected and mixed with 25 mL of 5 M renal and inguinal fat were significantly higher in the P group than in
NaOH solution and acetone. The processed samples were lyophilised the C group (p < .05); however, the weights of total body fat and
and stored for SCFA analysis. All measurements were performed in mesenteric fat were significantly lower in the B group than in the C
duplicate. group (p < .05). Overall, the weights of total body fat and selected
Chromatographic analysis was performed using the Trace Gas adipose tissue fat were significantly higher in the high-fat diet group
Chromatography (GC) system (Thermo Finnigan Trace GC, Milan, Italy), than in the normal diet group (p < .05).
equipped with a flame ionisation detector. A GC capillary column
(NukolTM 24107, Sigma-Aldrich) with a column size of 30 m, inner 3.2 | Lipid profile and safety
diameter of 0.25 mm, and a 0.25-lm-thick film coating was used.
We observed no significant differences in the plasma and liver levels of
Nitrogen was supplied as the carrier gas at a flowrate of 0.5 mL/min,
TC, HDLc, and LDLc among the groups at the end of the experiments;
with a split ratio of 1:10. The GC oven temperature was initially main-
however, the plasma TG level was significantly higher in the C group
tained at 1308C then increased 68C/min to 2008C and maintained for
than in the normal diet, B, and P groups (Table 3). Moreover, the daily
7.5 min. The temperature of the flame ionisation detector and injection
faecal weight was significantly higher in the B and M groups than in the
port was 2308C. The flowrates of hydrogen and air were 30 and
C group (p < .05 for both). Although blood urea nitrogen and creatinine
300 mL/min, respectively. The volume of the injected sample for GC
levels were significantly lower in the C group than in the normal diet
was 2 lL. Independently replicated determinations were performed
group, the levels were within the normal range. Liver and kidney function
three times for each sample.
indices after the intervention indicated that the diet was safe (Table 3).

2.6 | Statistical analysis 3.3 | Effects on gut microbiota


Data are expressed as the mean 6 standard error of the mean for each We observed that the numbers of E. coli CFUs were significantly higher
group. A p value of <.05 was considered statistically significant. One- in the rectal faeces of the treated groups than in those of the non-
way ANOVA was used to compare the mean values, and significant dif- treated groups (all p < .05). We also observed a significant increase in
ferences among groups were determined using Duncan’s multiple the number of Bifidobacterium CFUs in the rectal faeces of the M group
range test (PASW statistics 18, SPSS, Inc., Hong Kong). at the end of the treatments (Table 4).

3 | RESULTS 3.4 | SCFA levels


After 8 weeks, the levels of SCFAs, particularly acetate and propionate,
3.1 | Obesity status and growth of rats
in the caecal faeces were significantly higher in the M group than in
To investigate the obesity status and growth of rats during the the other treated groups (p ˂.05). However, no differences in SCFA lev-
experiment, we evaluated the dietary intake, body weight, and els were observed in the rectal faeces of the P, B, and M groups. The
selected adipose tissues (Table 2). We measured the body weight of SCFA levels in the rectal faeces of the P, B, and M groups were signifi-
all rats to evaluate the obesity status. We observed no differences in cantly higher than those of the C group (p < .05). After 8 weeks, com-
food intake and liver weight among the obese rat groups. After pared with other treated groups, the propionate and butyrate levels
treatment, the final body weight gain of the B group was signifi- were significantly higher in the B and M groups, respectively. More-
cantly lower than that of the C group (p < .05). The weights of peri- over, the levels of total SCFAs, acetate, propionate, and butyrate in the
LEE ET AL. | 5 of 9

TA BL E 3 Effects of 8-week consumption of L. plantarum and b-glucan on selected biochemical parameters

Groups/parameter N C P B M

Plasma
TC (mg/dL) 49.8 6 3.3 56.6 6 4.1 60.0 6 3.9 52.0 6 6.7 47.6 6 4.0
TG (mg/dL) 122 6 11 182 6 26*a 159 6 24ab 108 6 12b 112 6 10b
HDLc (mg/dL) 14.5 6 0.6 15.7 6 1.0 15.9 6 1.0 14.6 6 1.7 12.8 6 0.7
LDLc (mg/dL) 3.60 6 0.22 4.30 6 0.37 4.38 6 0.32 4.00 6 0.37 3.50 6 0.22
GOT (U/L) 99.9 6 12.7 91.5 6 6.1 91.5 6 7.1 86.1 6 4.2 96.3 6 17.9
GPT (U/L) 34.2 6 3.6 32.4 6 1.5 40.0 6 7.0 32.0 6 2.4 43.5 6 12.6
BUN (mg/dL) 11.7 6 0.5 9.7 6 0.3* 10.0 6 0.6 10.5 6 0.7 10.8 6 0.2
Creatinine (mg/dL) 0.40 6 0.01 0.37 6 0.01*a 0.38 6 0.01a 0.37 6 0.01b 0.40 6 0.01b
Liver
TC (mg/g) 3.09 6 0.87 2.25 6 0.16a 3.74 6 0.50b 3.24 6 0.67ab 2.98 6 0.62ab
TG (mg/g) 7.5 6 1.5 9.7 6 1.2a 13.3 6 2.2b 9.4 6 0.6a 10.2 6 0.9ab
Feces
Weight (g/d/rat) 2.93 6 0.84 3.87 6 0.58a 3.77 6 0.86a 7.29 6 2.07b 6.67 6 0.71b
Cholic acid (mmol/d/rat) 2.76 6 0.78 1.69 6 0.80 0.93 6 0.30 2.22 6 1.35 1.24 6 0.41
Water content (%) 0.38 6 0.09 0.33 6 0.05 0.28 6 0.06 0.29 6 0.09 0.24 6 0.09

Note. Data are expressed as the mean 6 SEM, n 5 8–10.


TC 5 total cholesterol; TG 5 triglyceride; HDLc 5 high-density lipoprotein cholesterol; LDLc 5 low-density lipoprotein cholesterol; GOT 5 glutamate
oxaloacetate transaminase; GPT 5 glutamate pyruvate transaminase; BUN 5 blood urea nitrogen. N 5 normal; C 5 control; P 5 Lactobacillus plantarum;
B 5 (1,3/1,4) b-glucan; M 5 Lactobacillus plantarum plus b-glucan.
The superscript symbol “*” means significantly different from the normal group (p < .05). Values in the same row with different superscript letters signif-
icantly differ at p < .05 by one-way ANOVA with least significant difference.

colonic mucin were higher in the P and M groups than in the other or a combination of both. The B and M groups exhibited the lowest
treated groups (p < .05) (Table 5). body weight gain and the highest daily faecal output (Kalra & Jood,
1998, 2000). However, the P and M groups showed significant body

3.5 | Selected hepatic lipogenic and lipolytic enzyme weight gain and body fat accumulation in specific adipose tissues (i.e.,
perirenal, inguinal, and mesenteric fat). The fat-regulating properties of
activities
L. plantarum P-8 have been reported in murine models (Kim et al.,
The activities of hepatic ACC, FAS, and CPT-1 were significantly higher 2014; Park et al., 2014): Lipolytic enzymes such as CPT-1 were regu-
in the B group than in the other treated groups (all p < .05; Figure 1). lated by L. plantarum LG42, resulting in a significant decrease in the fat
However, the LPL activity in the epididymal fat tissue significantly pad weight of epididymal adipose tissue (Park et al., 2014). Similar lipid
decreased in the P, B, and M groups (all p < .05; Figure 2). reduction is caused by Lactobacillus strains and occurs in hosts with
dysfunctional metabolisms (i.e., elevated serum insulin, TNFa, or IL-1b
4 | DISCUSSION levels) (Fak & Backhed, 2012; Wang et al., 2014). However, in our
study, we did not observe a similar decrease in white adipose tissue
In this study, we observed that body weight gain and body fat distribu- accumulation in the P and M groups. According to our study protocol,
tion differed among obese rats fed a high-fat diet supplemented with we fed most of our rats with the high-fat diet for 12 weeks, followed
barley b-glucan (primarily (1!3,1!4)-b-glucan; 90%), L. plantarum P-8, by the applicable treatment administration. After administration, the

TA BL E 4 Effects of 8-week consumption of L. plantarum and b-glucan on the gut microbiota in rectal feces

N C P B M

Endo Baseline 7.45 6 0.20 7.05 6 0.24 6.75 6 0.20 7.00 6 0.14 6.83 6 0.11
8th week 7.79 6 0.17 8.00 6 0.14* 7.85 6 0.35* 7.82 6 0.15* 7.21 6 0.21*
MRS Baseline 7.63 6 0.13 7.40 6 0.08 7.74 6 0.13 7.40 6 0.11 7.46 6 0.12
8th week 7.76 6 0.20 7.22 6 0.14 7.13 6 0.09* 7.57 6 0.15 7.60 6 0.24
BIM Baseline 7.57 6 0.22 7.33 6 0.18 7.41 6 0.14 7.30 6 0.16 7.17 60.10
8th week 7.67 6 0.20 7.28 6 0.11 7.24 6 0.15 7.37 6 0.09 7.69 6 0.13*

CDC Baseline 8.35 6 0.22 8.08 6 0.17 8.11 6 0.09 8.23 6 0.11 8.17 6 0.14
8th week 8.18 6 0.21 8.10 6 0.13 8.36 6 0.28 8.41 6 0.13 8.056 0.14

Note. Data are expressed as the mean 6 SEM, n 5 8–10. Values are expressed as log (colony-forming units per gram) of wet feces. Endo agar was used
for isolating Escherichia coli, MRS agar was used for isolating Lactobacillus, BIM agar was used for isolating Bifidobacterium, and CDC agar was used for
isolating total anaerobic bacteria.
N 5 normal; C 5 control; P 5 Lactobacillus plantarum; B 5 (1,3/1,4) b-glucan; M 5 Lactobacillus plantarum plus b-glucan.
The superscript symbol “*” means significantly different from the baseline in the same treatment (p < .05).
6 of 9 | LEE ET AL.

TA BL E 5 Levels of SCFAs in cecal and rectal feces and colon mucin of rats after administration

N C P B M

Cecum feces (mM/g wet)


Total SCFAs 49.5 6 3.7 58.9 6 5.0a 61.8 6 4.2a 53.2 6 2.9a 90.3 6 6.8b
Acetate 21.7 6 2.0 33.2 6 3.3*a 32.5 6 3.0a 32.6 6 1.4a 53.2 6 4.2b
Propionate 11.8 6 1.2 12.5 6 1.0a 13.1 6 0.8a 11.6 6 0.9a 20.9 6 1.9b
Butyrate 16.0 6 1.4 13.1 6 1.6ab 16.3 6 1.3b 9.0 6 1.6a 16.2 6 2.3bc
Rectal feces (mM/g wet)
Total SCFAs 11.8 6 1.0 14.4 6 1.5a 21.9 6 2.5b 23.4 6 2.3b 22.8 6 1.1b
Acetate 9.4 6 0.7 13.2 6 1.5a 20.3 6 2.3b 19.5 6 1.8b 19.5 6 1.1b
Propionate 1.0 6 0.1 0.7 6 0.1a 0.6 6 0.1a 1.7 6 0.2b 1.2 6 0.1a
Butyrate 1.5 6 0.2 0.5 6 0.0a 0.9 6 0.2ab 2.2 6 0.8bc 2.6 6 1.7c

Colon mucin (mM/g)


Total SCFAs 19.1 6 1.7 27.8 6 5.6a 46.2 6 9.8b 22.2 6 2.4a 13.6 6 1.7c
Acetate 16.0 6 1.4 22.6 6 4.5a 35.7 6 8.2b 17.1 6 1.9a 9.9 6 1.4c
Propionate 2.1 6 0.3 3.5 6 0.8a 7.0 6 1.5b 3.9 6 0.6a 2.5 6 0.3c
Butyrate 1.0 6 0.1 1.6 6 0.4a 3.5 6 0.7b 1.1 6 0.3a 1.2 6 0.2a

Note. Data are expressed as the mean 6 SEM, n 5 8–10.


SCFAs 5 short-chain fatty acids; N 5 normal; C 5 control; P 5 Lactobacillus plantarum; B 5 (1,3/1,4) b-glucan; M 5 Lactobacillus plantarum plus b-glucan.
The superscript letter “*” means significantly different from the normal group (p < .05). Values in the same row with different superscript letters signifi-
cantly differ at p < .05 by one-way ANOVA with least significant difference.

hepatic TC and TG levels did not differ between the control and normal strated multiple cholesterol-lowering effects not only in the structural
groups. These findings are inconsistent with those of other studies, trapping of bile acid but also in the elevation of CYP7A1 activity (Yang,
probably because this level of obesity (more than 10% of body weight) Kim, Lee, Lee, & Moon, 2003) and LDL receptor regulation (Watkins,
does not cause the onset of abnormal lipid metabolism in the liver 2004), which accelerate bile acid excretion and cholesterol utilisation,
(Abdul Kadir et al., 2015). High-fat diets are a major cause of obe- respectively. In this study, TC and faecal cholic acid levels did not differ
sity; but in mammals, despite long-term intake of a high-fat diet sig- among the treatment groups. However, serum TG levels were signifi-
nificantly increasing abdominal fat weight (Woods, Seeley, Rushing, cantly lower in the B and M groups than in the C group. No differences
D’Alessio, & Tso, 2003), biochemical parameters remain within nor- in other biochemical parameters were observed in the P group.
mal ranges (Table 3). In our study, the dietary intervention with b-glucan did not affect
Oat and barley b-glucans have been shown to reduce TC levels in food intake and white adipose tissue weight; nevertheless, a decreasing
people diagnosed with hypercholesterolaemia (FDA, 1997, 21 January) trend in food efficiency and white adipose tissue weight was observed.
and to increase bile acid excretion (Bae, Kim, Lee, & Lee, 2010; Ellegard By contrast, after the dietary intervention with L. plantarum, an increas-
& Andersson, 2007). High molecular weight b-glucan (1,200 kDa) has ing trend in body weight and food efficiency was observed. These
higher viscosity than low molecular weight b-glucan (400 kDa) and results are inconsistent with those of studies that used murine models
thus may trap more unabsorbed cholesterol and its derivatives (e.g., for examining the anti-obesity effects of L. plantarum (Lee, Paek, Lee,
bile salts) than low molecular weight b-glucan does. However, the Park, & Lee, 2007; Park et al., 2014; Yoo et al., 2013). Regarding stud-
lipid-lowering effects of high and low molecular weight b-glucans were ies of humans, the number of Firmicutes, such as Lactobacillus, have
reported to be similar (Keenan et al., 2007). Notably, b-glucan demon- been found to be higher in patients with obesity than in participants

F I G U R E 1 The percentage yields of different extracts obtained F I G U R E 2 Results of TPC and TFC of different extracts obtained
from the powdered roots of N. leucophylla from stems of N. leucophylla
LEE ET AL. | 7 of 9

with a normal weight; one study indicated that some Lactobacillus tary treatments (probiotic and prebiotics) may independently contrib-
strains may contribute to gut microbiota-induced obesity (Kadooka ute to weight gain and weight production. Moreover, some
et al., 2010; Million, Maraninchi, et al., 2012). The inconsistent results Lactobacillus spp. have modulating properties that release the fasting-
may be due to different degrees of weight gain caused by variation in induced adipocyte factor from intestinal cells, thereby inhibiting LPL
host-specific factors (i.e., animal species and host physical status), as activity and resulting in increased liver-derived TG storage (Backhed,
well as the different Lactobacillus species (Million, Angelakis, et al., Manchester, Semenkovich, & Gordon, 2007).
2012) and their secondary metabolites (i.e., SCFAs, t10, c12- Our study has a few limitations. First, although probiotics (e.g.,
conjugated linoleic acid, and cholic acid) (Byrne, Chambers, Morrison, & L. plantarum P-8) have been used to reduce weight gain in some murine
Frost, 2015; Lee et al., 2007). In our study, the P and M groups exhib- species, the subspecies of Lactobacillus involved in energy homeostasis
ited higher body weight gain and white adipose tissue accumulation in rats does not exhibit consistent properties in this respect. Second,
(inguinal and perirenal fat) than the C group did. The levels of total we did not quantify the changes in mucosal receptors and proinflam-
SCFAs, particularly butyrate, in rectal faeces and the colonic mucin matory factors. Although the results regarding bacteria-produced
were proportionally associated with weight gain and adipose tissue SCFAs indicated that SCFAs have a crucial role in energy homeostasis,
accumulation in the P and M groups. Several mechanisms through the expression levels of possible mucosal receptors (i.e., GPR41 and
which bacterial metabolites affect the regulation of energy homeostasis GRP43) and proinflammatory factors should be quantified. Additional

and obesity have been suggested (Erejuwa, Sulaiman, & Ab Wahab, studies investigating the inflammatory status (i.e., insulin, CRP, TNFa,

2014). Bacteria-produced SCFAs are thought to activate a series of and selected IL levels) are necessary. Third, we did not analyse the gly-

G-protein–coupled receptors (GPR41 and GRP43) in gut epithelial cells. caemic status of the rats, although the rats fed the high-fat diet for 8

The activation of SCFA-linked G-protein-coupled receptors, which con- weeks had normal liver and kidney functions and exhibited no extreme
changes in their liver lipid profiles. Notably, obesity-related metabolism
siderably increase energy harvest and deposition in hosts, can be attrib-
may trigger abnormal blood sugar and lipid regulation leading to the
uted to gut microbiota (Erejuwa et al., 2014). These results support the
onset of metabolic syndrome. Fourth, some activities of lipolytic and
alternative view that gut microbiota may have a key role in modulating
lipogenic enzymes were inconsistent among the treated groups (i.e.,
the harvest of energy from macronutrients, and may be a factor that
the P and B groups). The physical network of SCFAs and related energy
affects the metabolic status of the host, regulating additional energy
homeostasis parameters (i.e., intestinal mucosal barriers and carbohy-
expenditure and accumulation (Cani & Everard, 2016).
drate and lipid metabolic enzymes) could be interpreted as indicating
Regarding gut SCFA levels, we analysed the relationship among
that an unknown upstream neurotransmitter regulates macronutrient
SCFAs in caecal faeces, rectal faeces, and colonic mucin. Gut
metabolism. A well-designed study on L. plantarum should be con-
microbiota-produced SCFAs fulfil a relatively small portion of the
ducted to evaluate the effects of SCFAs on the activities of intestinal
energy requirement; however, studies have suggested that these
mucosal barriers and lipid metabolic enzymes.
metabolites may affect energy homeostasis (Moreno-Indias, Cardona,
In summary, our results indicate that administering b-glucan and L.
Tinahones, & Queipo-Ortuno, 2014; Moreno-Indias and Tinahones,
plantarum P-8 to obese rats produces different energy harvesting
2015). SCFAs regulate cell differentiation, activate intestinal neuro-
effects. b-Glucan, but not L. plantarum P-8, can increase the activities
transmitter release (de Theije et al., 2014; Zaibi et al., 2010), and regu-
of hepatic FAS, ACC, and CPT and reduce the activity of epididymal fat
late energy homeostasis through gluconeogenesis and lipogenesis in
LPL, resulting in decreased white adipose tissue accumulation in obese
specific organs such as the liver and muscles (Udayappan, Hartstra,
rats. However, b-glucan and L. plantarum P-8 demonstrated no syner-
Dallinga-Thie, & Nieuwdorp, 2014). In our study, we observed that the
gistic effects. Further evidence demonstrating the ability of SCFAs to
total SCFA levels in caecal and rectal faeces were significantly higher in
reduce body weight and white adipose tissue accumulation under a
the M group than in the C group. Butyrate levels in the colonic mucin
high-fat diet is necessary.
layer were significantly higher in the P and M groups than in the B
group. Therefore, we propose that SCFAs, particularly butyrate, pene-
ACKNOWLEDG MENTS
trate the intestinal epithelium and directly provide a signal that acti-
vates signalling cascade events, thereby enabling the cellular activities This research was partially supported by grants (NSC101-2320-B-

of lipogenic and lipolytic enzymes. The LPL activity of the M group was 038-020-MY2; NSC102-2313-B-038-003-MY2, and MOST103-

inversely correlated with gut SCFA levels. In addition, SCFA levels in 2320-B-038-012-MY3) from the Ministry of Science and Technol-
ogy, Taiwan.
the colonic mucin were higher in the P and M groups than in the other
treated groups. Furthermore, the B and M groups had lower SCFA lev-
els in the colonic mucin than other treated groups did. Although a trend CONFLIC T OF I NTE RE ST
of increasing SCFA levels in the colon mucin and of increasing hepatic The authors declared that they have no conflict of interest.
ACC and FAS activities was observed in the P and M groups, the SCFA
levels produced by 109 CFU of L. plantarum P-8 did not reach the RE FE RE NCE S
threshold for activating enzymes involved in energy homeostasis. Thus, Abdul Kadir, N. A., Rahmat, A., & Jaafar, H. Z. (2015). Protective effects
SCFAs in various locations (faeces and mucin) and under different die- of Tamarillo (Cyphomandra betacea) extract against high fat diet
8 of 9 | LEE ET AL.

induced obesity in Sprague-Dawley rats. Journal of Obesity, 2015, Kalra, S., & Joad, S. (2000). Effect of dietary barley beta-glucan on
846041. cholesterol and lipoprotein fractions in rats. Journal of Cereal Science,
Arena, M. P., Caggianiello, G., Fiocco, D., Russo, P., Torelli, M., Spano, G., 31, 141–145.
& Capozzi, V. (2014). Barley beta-glucans-containing food enhances Keenan, J. M., Goulson, M., Shamliyan, T., Knutson, N., Kolberg, L., &
probiotic performances of beneficial bacteria. International Journal of Curry, L. (2007). The effects of concentrated barley beta-glucan on
Molecular Sciences, 15, 3025–3039. blood lipids in a population of hypercholesterolaemic men and
Backhed, F., Manchester, J. K., Semenkovich, C. F., & Gordon, J. I. women. British Journal of Nutrition, 97, 1162–1168.
(2007). Mechanisms underlying the resistance to diet-induced obesity Kim, Y., Yoon, S., Lee, S. B., Han, H. W., Oh, H., Lee, W. J., & Lee, S. M.
in germ-free mice. Proceedings of the National Academy of Sciences of (2014). Fermentation of soy milk via Lactobacillus plantarum improves
the United States of America, 104, 979–984. dysregulated lipid metabolism in rats on a high cholesterol diet. PLoS
Bae, I. Y., Kim, S. M., Lee, S., & Lee, H. G. (2010). Effect of enzymatic One, 9, e88231.
hydrolysis on cholesterol-lowering activity of oat beta-glucan. New Kumar, R., Grover, S., & Batish, V. K. (2011). Hypocholesterolaemic effect
Biotechnology, 27, 85–88. of dietary inclusion of two putative probiotic bile salt hydrolase-
Byrne, C. S., Chambers, E. S., Morrison, D. J., & Frost, G. (2015). The role producing Lactobacillus plantarum strains in Sprague-Dawley rats.
of short chain fatty acids in appetite regulation and energy homeosta- British Journal of Nutrition, 105, 561–573.
sis. International Journal of Obesity (London), 39, 1331–1338. Lee, K., Paek, K., Lee, H. Y., Park, J. H., & Lee, Y. (2007). Antiobesity
Cani, P. D., & Everard, A. (2016). Talking microbes: When gut bacteria effect of trans-10,cis-12-conjugated linoleic acid-producing Lactoba-
interact with diet and host organs. Molecular Nutrition & Food cillus plantarum PL62 on diet-induced obese mice. Journal of Applied
Research, 60, 58–66. Microbiology, 103, 1140–1146.

Carlson, J. J., Eisenmann, J. C., Norman, G. J., Ortiz, K. A., & Young, P. C. Ley, R. E., Backhed, F., Turnbaugh, P., Lozupone, C. A., Knight, R. D., &
(2011). Dietary fiber and nutrient density are inversely associated Gordon, J. I. (2005). Obesity alters gut microbial ecology. Proceedings
with the metabolic syndrome in US adolescents. The Journal of the of the National Academy of Sciences of the United States of America,
American Dietetic Association, 111, 1688–1695. 102, 11070–11075.

de Theije, C. G., Wopereis, H., Ramadan, M., van Eijndthoven, T., Lam- Million, M., Angelakis, E., Paul, M., Armougom, F., Leibovici, L., & Raoult,
bert, J., Knol, J., . . . Oozeer, R. (2014). Altered gut microbiota and D. (2012a). Comparative meta-analysis of the effect of Lactobacillus
activity in a murine model of autism spectrum disorders. Brain, species on weight gain in humans and animals. Microbial Pathogenesis,
Behavior, and Immunity, 37, 197–206. 53, 100–108.

Druart, C., Dewulf, E. M., Cani, P. D., Neyrinck, A. M., Thissen, J. P., & Million, M., Maraninchi, M., Henry, M., Armougom, F., Richet, H., Carrieri,
Delzenne, N. M. (2014). Gut microbial metabolites of polyunsatu- P., . . . Raoult, D. (2012). Obesity-associated gut microbiota is
rated fatty acids correlate with specific fecal bacteria and serum enriched in Lactobacillus reuteri and depleted in Bifidobacterium ani-
markers of metabolic syndrome in obese women. Lipids, 49, 397– malis and Methanobrevibacter smithii. International Journal of Obesity
402. (London), 36, 817–825.
Ellegard, L., & Andersson, H. (2007). Oat bran rapidly increases bile acid Moreno-Indias, I., Cardona, F., Tinahones, F. J., & Queipo-Ortuno, M. I.
excretion and bile acid synthesis: An ileostomy study. European Jour- (2014). Impact of the gut microbiota on the development of obesity
nal of Clinical Nutrition, 61, 938–945. and type 2 diabetes mellitus. Frontiers in Microbiology, 5, 190.
Erejuwa, O. O., Sulaiman, S. A., & Ab Wahab, M. S. (2014). Modulation Moreno-Indias, I., & Tinahones, F. J. (2015). Impaired adipose tissue
of gut microbiota in the management of metabolic disorders: The expandability and lipogenic capacities as ones of the main causes of
prospects and challenges. International Journal of Molecular Sciences, metabolic disorders. Journal of Diabetes Research, 2015, 970375.
15, 4158–4188. Nielsen, T. S., Jensen, B. B., Purup, S., Jackson, S., Saarinen, M., Lyra, A., . . . Knud-
Ezz El-Arab, A. M. (2009). A diet rich in leafy vegetable fiber improves sen, K. E. B. (2016). A search for synbiotics: Effects of enzymatically modified
cholesterol metabolism in high-cholesterol fed rats. Pakistan Journal arabinoxylan and Butyrivibrio fibrisolvens on short-chain fatty acids in the
of Biological Sciences, 12, 1299–1306. cecum content and plasma of rats. Food & Function, 7, 1839–1848.
Fak, F., & Backhed, F. (2012). Lactobacillus reuteri prevents diet-induced Notarnicola, M., Miccolis, A., Tutino, V., Lorusso, D., & Caruso, M. G.
obesity, but not atherosclerosis, in a strain dependent fashion in (2012). Low levels of lipogenic enzymes in peritumoral adipose tissue
Apoe2/2 mice. PLoS One, 7, e46837. of colorectal cancer patients. Lipids, 47, 59–63.
FDA (1997). FDA Talk Paper. FDA allows whole oat foods to make Numa, S., Bortz, W. M., & Lynen, F. (1965). Regulation of fatty acid syn-
health claim on reducing the risk of heart disease, http://www.cfsan. thesis at the acetyl-CoA carboxylation step. Advances in Enzyme Reg-
fda.gov/~lrd/tpoats.html (accessed January 21, 1997). ulation, 3, 407–423.
Ghibaudi, L., Cook, J., Farley, C., van Heek, M., & Hwa, J. J. (2002). Fat Ogden, C. L., Carroll, M. D., & Flegal, K. M. (2014). Prevalence of obesity
intake affects adiposity, comorbidity factors, and energy metabolism in the United States. JAMA, 312, 189–190.
of Sprague-Dawley rats. Obesity Research, 10, 956–963. Park, J. E., Oh, S. H., & Cha, Y. S. (2014). Lactobacillus plantarum LG42 iso-
Hu, F. B. (2003). Plant-based foods and prevention of cardiovascular dis- lated from gajami sik-hae decreases body and fat pad weights in diet-
ease: An overview. The American Journal of Clinical Nutrition, 78, induced obese mice. Journal of Applied Microbiology, 116, 145–156.
544s–551s. Salazar, N., Dewulf, E. M., Neyrinck, A. M., Bindels, L. B., Cani, P. D., Mahil-
Kadooka, Y., Sato, M., Imaizumi, K., Ogawa, A., Ikuyama, K., Akai, Y., . . . Tsu- lon, J., . . . Delzenne, N. M. (2015). Inulin-type fructans modulate intestinal
chida, T. (2010). Regulation of abdominal adiposity by probiotics (Lactoba- Bifidobacterium species populations and decrease fecal short-chain fatty
cillus gasseri SBT2055) in adults with obese tendencies in a randomized acids in obese women. Clinical Nutrition, 34, 501–507.
controlled trial. European Journal of Clinical Nutrition, 64, 636–643. Small, G. M., Burdett, K., & Connock, M. J. (1985). A sensitive spectro-
Kalra, S., & Jood, S. (1998). Biological evaluation of protein quality of photometric assay for peroxisomal acyl-CoA oxidase. Biochemical
barley. Food Chemistry, 61, 35–39. Journal, 227, 205–210.
LEE ET AL. | 9 of 9

Tilg, H., & Moschen, A. R. (2014). Microbiota and diabetes: An evolving Wu, H., Tremaroli, V., & Backhed, F. (2015). Linking microbiota to human
relationship. Gut, 63, 1513–1521. diseases: A systems biology perspective. Trends in Endocrinology &
Tilg, H., Moschen, A. R., & Kaser, A. (2009). Obesity and the microbiota. Metabolism, 26, 758–770.
Gastroenterology, 136, 1476–1483. Yang, J. L., Kim, Y. H., Lee, H. S., Lee, M. S., & Moon, Y. K. (2003). Barley
Turnbaugh, P. J., Hamady, M., Yatsunenko, T., Cantarel, B. L., Duncan, A., beta-glucan lowers serum cholesterol based on the up-regulation of
Ley, R. E., . . . Gordon, J. I. (2009). A core gut microbiome in obese cholesterol 7alpha-hydroxylase activity and mRNA abundance in
and lean twins. Nature, 457, 480–484. cholesterol-fed rats. The Journal of Nutritional Science and Vitaminol-
ogy (Tokyo), 49, 381–387.
Turnbaugh, P. J., Ley, R. E., Mahowald, M. A., Magrini, V., Mardis, E. R.,
Yoo, S. R., Kim, Y. J., Park, D. Y., Jung, U. J., Jeon, S. M., Ahn, Y. T., . . .
& Gordon, J. I. (2006). An obesity-associated gut microbiome with
Choi, M. S. (2013). Probiotics L. plantarum and L. curvatus in combi-
increased capacity for energy harvest. Nature, 444, 1027–1031.
nation alter hepatic lipid metabolism and suppress diet-induced obe-
Udayappan, S. D., Hartstra, A. V., Dallinga-Thie, G. M., & Nieuwdorp, M. sity. Obesity (Silver Spring), 21, 2571–2578.
(2014). Intestinal microbiota and faecal transplantation as treatment
Zaibi, M. S., Stocker, C. J., O’dowd, J., Davies, A., Bellahcene, M., Caw-
modality for insulin resistance and type 2 diabetes mellitus. Clinical &
thorne, M. A., . . . Arch, J. R. (2010). Roles of GPR41 and GPR43 in
Experimental Immunology, 177, 24–29.
leptin secretory responses of murine adipocytes to short chain fatty
Wang, L., Zhang, J., Guo, Z., Kwok, L., Ma, C., Zhang, W., . . . Zhang, H. acids. FEBS Letters, 584, 2381–2386.
(2014). Effect of oral consumption of probiotic Lactobacillus planta-
Zhong, Y., Marungruang, N., Fak, F., & Nyman, M. (2015). Effects of two
rum P-8 on fecal microbiota, SIgA, SCFAs, and TBAs of adults of dif-
whole-grain barley varieties on caecal SCFA, gut microbiota and
ferent ages. Nutrition, 30, 776–783.e771.
plasma inflammatory markers in rats consuming low- and high-fat
Watkins, L. O. (2004). Epidemiology and burden of cardiovascular dis- diets. British Journal of Nutrition, 113, 1558–1570.
ease. Clinical Cardiology, 27, III2–III6.
WHO (2016). Obesity and overweight. World Health Organization Fact
sheet, http://www.who.int/mediacentre/factsheets/fs311/en/.
How to cite this article: Lee H-C, Yu W-T, Guo Y-R, Huang S-Y.
Wong, J. M., de Souza, R., Kendall, C. W., Emam, A., & Jenkins, D. J.
b-Glucan, but not Lactobacillus plantarum P-8, inhibits lipid
(2006). Colonic health: Fermentation and short chain fatty acids.
accumulation through selected lipid metabolic enzymes in
Journal of Clinical Gastroenterology, 40, 235–243.
obese rats. J Food Biochem. 2017;41:e12336. https://doi.org/
Woods, S. C., Seeley, R. J., Rushing, P. A., D’alessio, D., & Tso, P. (2003).
A controlled high-fat diet induces an obese syndrome in rats. Journal 10.1111/jfbc.12336
of Nutrition, 133, 1081–1087.

You might also like