ArticlePDF Available

Effect of Oat β-Glucan on Affective and Physical Feeling States in Healthy Adults: Evidence for Reduced Headache, Fatigue, Anxiety and Limb/Joint Pains

MDPI
Nutrients
Authors:
  • INQUIS Clinical Research, Ltd

Abstract and Figures

The gastrointestinal (GI) side-effects of dietary fibers are recognized, but less is known about their effects on non-GI symptoms. We assessed non-GI symptoms in a trial of the LDL-cholesterol lowering effect of oat β-glucan (OBG). Participants (n = 207) with borderline high LDL-cholesterol were randomized to an OBG (1 g OBG, n = 104, n = 96 analyzed) or Control (n = 103, n = 95 analyzed) beverage 3-times daily for 4 weeks. At screening, baseline, 2 weeks and 4 weeks participants rated the severity of 16 non-GI symptoms as none, mild, moderate or severe. The occurrence and severity (more or less severe than pre-treatment) were compared using chi-squared and Fisher’s exact test, respectively. During OBG treatment, the occurrence of exhaustion and fatigue decreased versus baseline (p < 0.05). The severity of headache (2 weeks, p = 0.032), anxiety (2 weeks p = 0.059) and feeling cold (4 weeks, p = 0.040) were less on OBG than Control. The severity of fatigue and hot flashes at 4 weeks, limb/joint pain at 2 weeks and difficulty concentrating at both times decreased on OBG versus baseline. High serum c-reactive-protein and changes in c-reactive-protein, oxidized-LDL, and GI-symptom severity were associated with the occurrence and severity of several non-GI symptoms. These data provide preliminary, hypothesis-generating evidence that OBG may reduce several non-GI symptoms in healthy adults.
Content may be subject to copyright.
nutrients
Article
Effect of Oat β-Glucan on Affective and Physical Feeling States
in Healthy Adults: Evidence for Reduced Headache, Fatigue,
Anxiety and Limb/Joint Pains
Thomas M. S. Wolever 1,*, Maike Rahn 2,† , El Hadji Dioum 2, Alexandra L. Jenkins 1, Adish Ezatagha 1,
Janice E. Campbell 1and YiFang Chu 2


Citation: Wolever, T.M.S.; Rahn, M.;
Dioum, E.H.; Jenkins, A.L.; Ezatagha,
A.; Campbell, J.E.; Chu, Y. Effect of
Oat β-Glucan on Affective and
Physical Feeling States in Healthy
Adults: Evidence for Reduced
Headache, Fatigue, Anxiety and
Limb/Joint Pains. Nutrients 2021,13,
1534. https://doi.org/10.3390/
nu13051534
Academic Editors: Seiichiro Aoe,
Tatsuya Morita and Naohito Ohno
Received: 12 March 2021
Accepted: 29 April 2021
Published: 1 May 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Formerly GI Labs, INQUIS Clinical Research, Ltd., Toronto, ON M5C 2N8, Canada;
alexandrajenkins@inquis.com (A.L.J.); aezatagha@inquis.com (A.E.); jcampbell@inquis.com (J.E.C.)
2Quaker Oats Center of Excellence, PepsiCo R&D Nutrition, Barrington, IL 60010, USA;
mrinny96@gmail.com (M.R.); ElHadji.Dioum@pepsico.com (E.H.D.); yifang.chu@pepsico.com (Y.C.)
*Correspondence: twolever@inquis.com; Tel.: +1-416-861-9177
Current address: DSM Nutritional Products, North America Nutrition Science and Advocacy,
Parsippany, NJ 07054, USA.
Abstract:
The gastrointestinal (GI) side-effects of dietary fibers are recognized, but less is known about
their effects on non-GI symptoms. We assessed non-GI symptoms in a trial of the LDL-cholesterol
lowering effect of oat
β
-glucan (OBG). Participants (n= 207) with borderline high LDL-cholesterol
were randomized to an OBG (1 g OBG, n= 104, n= 96 analyzed) or Control (n= 103, n= 95 analyzed)
beverage 3-times daily for 4 weeks. At screening, baseline, 2 weeks and 4 weeks participants rated
the severity of 16 non-GI symptoms as none, mild, moderate or severe. The occurrence and severity
(more or less severe than pre-treatment) were compared using chi-squared and Fisher’s exact test,
respectively. During OBG treatment, the occurrence of exhaustion and fatigue decreased versus
baseline (p< 0.05). The severity of headache (2 weeks, p= 0.032), anxiety (2 weeks p= 0.059) and
feeling cold (4 weeks, p= 0.040) were less on OBG than Control. The severity of fatigue and hot
flashes at 4 weeks, limb/joint pain at 2 weeks and difficulty concentrating at both times decreased
on OBG versus baseline. High serum c-reactive-protein and changes in c-reactive-protein, oxidized-
LDL, and GI-symptom severity were associated with the occurrence and severity of several non-GI
symptoms. These data provide preliminary, hypothesis-generating evidence that OBG may reduce
several non-GI symptoms in healthy adults.
Keywords:
randomized clinical trial; humans; symptoms; gastrointestinal tract; musculo-skeletal
system; oats; oatmeal; dietary fiber; beta-glucan
1. Introduction
To protect against malnutrition and non-communicable diseases global nutrition rec-
ommendations include advice to consume more whole grains to help maintain an adequate
intake of dietary fiber [
1
]. Oats are a whole grain containing the soluble dietary fiber
β
-glucan [
2
]. Oat
β
-glucan (OBG) reduces serum LDL-cholesterol [
3
,
4
] and postprandial
glycemic responses [
5
], physiological effects associated with reduced risk for cardiovas-
cular disease [
6
] and type 2 diabetes [
7
], respectively. Dietary fibers such as OBG are not
absorbed in the small intestine and reach the colon where they increase the mass of the
colonic contents and may be partly or completely fermented by colonic microbiota, effects
which have desirable and undesirable consequences as explained below.
The undesirable side-effects of dietary fibers include abdominal bloating and pain,
flatus and diarrhea which, although transitory in nature, may limit the acceptability of high
fiber foods [
8
]. Consistent with this were our findings in healthy subjects who consumed
3 g/day OBG or Control for 4 weeks. After 2 weeks, flatulence and abdominal discomfort
Nutrients 2021,13, 1534. https://doi.org/10.3390/nu13051534 https://www.mdpi.com/journal/nutrients
Nutrients 2021,13, 1534 2 of 14
had increased from baseline on both the OBG and Control treatments; but by 4 weeks
these symptoms had begun to return to baseline [
9
]. In a cross-sectional study of adults
with coeliac disease, higher long-term oat intake correlated with lower gastrointestinal
symptom scores [10].
The desirable effects of increased fiber intake include increased stool bulk which
may be beneficial for conditions such as constipation, irritable bowel syndrome [
11
] and
diverticulosis [
12
] although not all types of fiber are equally effective. Oats and oat bran
appear to increase stool weight per gram of dietary fiber to a similar extent as other
sources of fiber [
13
]. Oats also decrease fecal pH (a marker of increased short-chain
fatty acid (SCFA) production) and increase the growth of beneficial gut microbiota [
14
].
Plausible mechanisms have been proposed whereby the stimulation of SCFA production
and alteration of the colonic microbiome can influence systemic oxidative stress and
inflammation and thereby influence both physical and mental functioning [
15
17
]. In this
context, it is of interest that higher self-reported fiber intake was associated with higher
positive affect in healthy children [
18
] and, in a randomized, cross-over study, consumption
of a high fiber breakfast cereal for 2 weeks was associated with less fatigue compared to
control in healthy adults [
19
]. We found that participants with type 2 diabetes treated
by diet alone who consumed a low glycemic index diet containing 37 g/day fiber for
1 year experienced less severe headaches, less severe pains in joints or limbs and less
severe gloomy thoughts than those on the lower fiber (21–23 g/day) diets [
20
]. OBG has
been shown to enhance the endurance capacity of rats and influence serum metabolites in
such a way as to suggest anti-fatigue properties [
21
], but there is a paucity of information
about the effect of OBG on non-gastrointestinal symptoms in humans. To address this,
we assessed a panel of non-gastrointestinal symptoms as tertiary objectives in a study the
primary purpose of which was to determine if a novel oat product containing OBG would
reduce serum LDL-cholesterol [9].
2. Materials and Methods
We performed a randomized, double-blind, placebo controlled, parallel arm design
clinical trial at INQUIS Clinical Research, a contract research organization. The protocol
was approved by the Western Institutional Review Board
®
(Puyallup, WA, USA) and all
participants provided informed consent by signing the IRB approved consent form. The
study was registered at www.clinicaltrials.gov (accessed on 11 April, 2019) with identifier:
NCT03911427.
Participants were healthy males and non-pregnant, non-lactating females without
diabetes, aged 18–65 year, with fasting calculated LDL-cholesterol between 3.00 and
5.00 mmol/L, inclusive, and BMI between 18.5 to <40 kg/m
2
. Details of the inclu-
sion/exclusion criteria can be found in Supplementary Information.
After signing the consent form, participants were screened over a period of 2–4 weeks
involving 2 visits to the clinic. First they answered questions about their medical and
drug history and had their height, weight and blood pressure measured, then they gave a
fasting blood sample and were given instructions about how to record a 3-day diet record
(3DDR) [
22
24
]. Participants who were eligible based on the results of the screening blood
sample were contacted and asked to start recording a 3DDR and attend at the clinic to
review the 3DDR and fill out the symptoms questionnaire (second screening visit, Screen 2).
To avoid the potential confounding effects of high intakes of saturated fat or dietary fiber
on the results, participants were excluded if their intake of saturated fat was
15% of
energy or they were consuming >14 g/1000 kcal dietary fiber. Eligible participants were
contacted by telephone or e-mail, advised to follow their usual dietary and exercise habits
and to refrain from consuming oat, barley and psyllium products for the duration of the
study and an appointment was made for the baseline visit.
Nutrients 2021,13, 1534 3 of 14
2.1. Study Visits
Eligible participants visited the clinic after 10–14 h overnight fasts at baseline (week 0)
and at 2 and 4 weeks, having been asked to avoid alcohol and unusual levels of physical
activity and food intake for 24 h before each visit. At each visit medications and adverse
events were reviewed, weight and blood pressure were measured, a fasting blood sample
obtained and a symptoms questionnaire filled out. At baseline, participants were randomly
assigned to either the Test or Control treatment. After the blood sample at each visit
participants consumed the first daily treatment sachet and were given a snack or light
breakfast. At baseline and week 2 participants were provided with a sufficient supply
of treatment sachets to last until the next visit and a shaker cup and whisk with which
to mix their assigned treatment and were given a study diary in which to record sachet
consumption. Compliance was checked at week 2 and week 4 by counting unused sachets
and, at week 2, participants were provided with forms and instructions to record another
3DDR before the next visit. Symptom questionnaires were filled out during each of the
visits at Screen 2, Baseline, 2 weeks and 4 weeks.
2.2. Interventions
Interventions were provided in color-coded sachets: the Test intervention consisted
of an oat ingredient delivering 1 g of
β
-glucan sachet; the Control intervention was a rice
milk powder containing 0 g
β
-glucan. Each sachet contained 0.9 g fat (0 g saturated),
1.9 g protein, ~14 g available carbohydrate and 1.9 g (Test) or 0.3 g (Control) dietary fiber.
Detailed nutrition information is given in Supplementary Table S1. Each sachet was mixed
with 8 oz (240 mL) of cold water, shaken for 30 sec and consumed immediately. The Test
and Control beverages had similar look, taste and smell. Subjects were asked to consume
3 sachets daily on 3 separate occasions separated by at least 3 h and preferably immediately
before or within 10 min of each main meal (breakfast, lunch and dinner).
2.3. Symptoms Questionnaire
Participants were asked to rate the severity of each of 27 symptoms (11 gastrointestinal
(GI) symptoms and 16 non-GI symptoms) they experienced over the previous 2 weeks on a
4-point scale: none = 0, mild = 1, moderate = 2 or severe = 3. We used this questionnaire in
2 previous studies in patients with diabetes [
20
,
22
,
23
]. The results for all 16 GI symptoms
have been reported elsewhere [
9
] but the sum of the scores for the 5 most prevalent (major)
GI symptoms (flatulence, diarrhea, constipation, abdominal distention and abdominal
pain) are included below. Of the 16 non-GI symptoms the following 4 were experienced by
a maximum of less than 10% of participants at any visit (baseline, 2 weeks and 4 weeks,
respectively) and the results are not presented here: Increased appetite (6%, 9% and 5%),
Palpitation/Throbbing of heart (4%, 6%, 4%), Balance disturbances (5%, 3%, 5%) and
Numbness/burning/itching hands feet (8%, 4%, 5%). The remaining 12 symptoms were
as follows: Headache, Fatigue, Lack of appetite, Tend to become exhausted (Exhaustion),
Feelings of anxiety, Lack of energy, Pains in joints or limbs (Limb/joint pain), Diminished
ability to concentrate (Reduced ability to concentrate), Feeling cold, Hot flashes/sensation
of rising heat (Hot flashes), Gloomy thoughts and Inner tension.
2.4. Data Analysis, Management and Calculations
Blood samples for blood lipids and lipoproteins, glucose and insulin were collected
at Baseline, 2 weeks and 4 weeks, and for glycated albumin, high sensitivity c-reactive
protein (CRP) and oxidized LDL (oxLDL) at Baseline and 4 weeks and the results presented
elsewhere [
9
]. However, results for CRP (analyzed by LifeLabs, Inc., Mississauga, ON,
USA) and oxLDL (analyzed by ELISA kit, catalog #30-7810, Alpco Diagnostics, Salem, NH,
USA) are included here because of their potential association with affective and physical
symptoms. Each 3DDR was reviewed with the participant by a dietitian or nutritionist
and subsequently analyzed for nutrient content (ESHA Food Processor, ESHA Research,
Nutrients 2021,13, 1534 4 of 14
Salem, OR, USA). The nutrients contained in the treatment sachets were added to those
from foods recorded on the 3DDR based on the sachet consumption counts
2.5. Power Analysis
The sample size calculation assumed that the SD of the change in LDL-cholesterol
between baseline and 4 weeks would be 0.455 mmol/L [
24
]. Using this SD, n= 90 subjects
per arm provides 80% power to detect an 0.19 mmol/L difference (5%) in LDL-cholesterol
change between the test and control arms. To improve the likelihood that at least n= 180
would complete the study we allowed for to 15% dropouts and enrolled 207 subjects.
2.6. Statistical Analysis
Included in this analysis are all 191 subjects who were randomized and completed the
study with no protocol violations and with 90% compliance (76/84 sachets over 4 weeks)
based on the sachet count. The highly skewed distributions of CRP and oxLDL values were
normalized by log transformation. Categorical measures were summarized as frequencies.
To determine whether the prevalence of symptoms during treatment differed from that
at the Baseline visit, and differed between treatments, the proportions of subjects who
experienced any symptom (mild, moderate or severe) were compared using the chi-square
test. To determine if the severity of symptoms during treatment differed from those before
treatment, the ratio of the number of subjects in each treatment group whose scores at
2 weeks and 4 weeks were greater (more severe, M) or less (less severe, L) than the median
score before treatment (defined as the median of the scores at the second screening (Scr2)
and Baseline visits) were compared to an expected ratio of 1:1 by chi-square test. To
determine if the change in severity of symptoms differed between treatments, the ratios of
L:M on Test and Control were compared by Fisher’s exact test [
20
]. Similarly, the severity
of symptoms in participants whose serum CRP, serum oxLDL and the sum of the scores for
5 major GI symptoms (flatulence, diarrhea, constipation, abdominal bloating, abdominal
discomfort) increased versus those in whom they did not increase during treatment.
3. Results
Recruitment began in April 2019 and the last subject visit occurred in February, 2020.
The recruitment questionnaire was completed by 2607 individuals of whom 1690 were
invited for screening; 538 attended Screen 1 (243 ineligible, 15 lost and 16 withdrawn),
264 attended for Screen 2 (7 excluded for SFA intake > 15% en, 10 excluded for dietary fiber
intake >14 g/1000 kcal, 8 lost and 15 withdrawn) and 224 were randomized (17 withdrawn,
103 received Control and 104 received Test). Four (4) Control participants were withdrawn
early for non-compliance (n= 3) or a serious adverse event (not related to treatment,
n= 1) and 4 participants were withdrawn after completion for non-compliance (n= 2) or
antibiotic use (n= 2) leaving n= 95 Control participants who completed the study per
protocol. Similarly, 3 Test participants were withdrawn early for antibiotic use (n= 1) or
scheduling difficulties (n= 2) and 5 were withdrawn after completion for non-compliance
(n= 2), antibiotic use (n= 2) or insulin use (n= 1) leaving n= 96 Test participants who
completed the study per protocol. The study flow chart and further details are given
elsewhere [9].
The per-protocol population consisted of 72 males and 119 females of whom 102
were Caucasian, 39 South Asian, 14 African, 14 Hispanic, 8 East Asian, 6 South-east Asian,
4 West Asian, 3 mixed and 1 Indigenous; participant sex and ethnicity did not differ
significantly on Test vs. Control. Participants were aged (mean
±
SD) 47.6
±
11.4 years,
BMI 27.9
±
4.6 kg/m
2
(BMI 25.0–<30, n= 191; BMI 30.0–<35, n= 132; BMI 35.0–<40, n= 43),
total cholesterol 5.83
±
0.70 mmol/L, triglycerides 1.46
±
0.69 mmol/L, HDL-cholesterol
1.44
±
0.41, LDL-cholesterol 3.73
±
0.50, fasting glucose 4.95
±
0.51 with no significant
differences between Test and Control
Mean energy intake increased from baseline on Test and Control by approximately
the amount of energy the sachets contained (Figure 1). Sugars intakes, increased to an
Nutrients 2021,13, 1534 5 of 14
equivalent extent on both treatments. Starch intake did not change significantly on Test
but increased from baseline on Control. Dietary fiber intake increased during treatment on
Test by approximately the amount in the Test sachets and the difference was significant
compared both to baseline and to Control (Figure 1). Total fat and protein intakes increased
from baseline on Control, but not Test; alcohol intake decreased from baseline on Test.
Nutrients 2021, 13, x FOR PEER REVIEW 5 of 14
1.44 ± 0.41, LDL-cholesterol 3.73 ± 0.50, fasting glucose 4.95 ± 0.51 with no significant
differences between Test and Control
Mean energy intake increased from baseline on Test and Control by approximately
the amount of energy the sachets contained (Figure 1). Sugars intakes, increased to an
equivalent extent on both treatments. Starch intake did not change significantly on Test
but increased from baseline on Control. Dietary fiber intake increased during treatment
on Test by approximately the amount in the Test sachets and the difference was significant
compared both to baseline and to Control (Figure 1). Total fat and protein intakes
increased from baseline on Control, but not Test; alcohol intake decreased from baseline
on Test.
Test Cont Test Cont Test Cont Test Cont
0
500
1000
1500
2000
0
10
20
30
40
Sugars
Starch
Fat
Protein
Alcohol
Dietary Fiber
Screen 2 Week 4
**
*
*
*
*
*
p<0.05
Screen 2 Week 4
Figure 1. Macronutrient and dietary fiber intakes during the trial. Bars represent mean ± SEM
intakes (expressed in Kcal for sugars, starch, fat, protein and alcohol) and grams for dietary fiber
before treatment (dietary records obtained at the second screening visit; Screen 2) and during
treatment (Week 4) for n = 96 on Test and n = 95 on Control (Cont). Error bars go down for sugars,
starch, fat, protein and dietary fiber and up for alcohol. The only significant difference between
Test and Control is for fiber. * Asterisks below the error bars indicate a significant difference from
Screen 2 for the respective nutrient, except for alcohol the asterisk is above the error bar, (p < 0.05)
by paired t-test. ƒ Significant difference in energy intake between Screen 2 and Week 4 (p < 0.05) by
paired t-test.
3.1. Gastrointestinal (GI) Symptoms
The occurrence and severity of GI symptoms are presented in detail elsewhere [9].
One or more major GI symptom (flatulence, diarrhea, constipation, abdominal distention, and
abdominal pain) was present in 36% of Test and 28% of Control participants at baseline (ns);
the prevalence increased to 61% and 52%, respectively, at week 2 and fell to 47% and 42%
by week 4 (differences between Test and Control were not significant). Furthermore, the
sum of the scores for the 5 major GI symptoms did not differ between Test and Control,
respectively, at any visit with mean ± SD as follows: screen 2, 0.95 ± 1.48 vs. 0.81 ± 1.55;
baseline, 0.77 ± 1.39 vs. 0.53 ± 1.29; week 2, 1.67 ± 2.12 vs. 1.45 ± 2.09; week 4, 1.20 ± 1.83 vs.
0.91 ± 1.39.
Figure 1.
Macronutrient and dietary fiber intakes during the trial. Bars represent mean
±
SEM intakes
(expressed in Kcal for sugars, starch, fat, protein and alcohol) and grams for dietary fiber before
treatment (dietary records obtained at the second screening visit; Screen 2) and during treatment
(Week 4) for n= 96 on Test and n= 95 on Control (Cont). Error bars go down for sugars, starch, fat,
protein and dietary fiber and up for alcohol. The only significant difference between Test and Control
is for fiber. * Asterisks below the error bars indicate a significant difference from Screen 2 for the
respective nutrient, except for alcohol the asterisk is above the error bar, (p< 0.05) by paired t-test.
ƒ
Significant difference in energy intake between Screen 2 and Week 4 (p< 0.05) by paired t-test.
3.1. Gastrointestinal (GI) Symptoms
The occurrence and severity of GI symptoms are presented in detail elsewhere [
9
].
One or more major GI symptom (flatulence, diarrhea, constipation, abdominal distention,
and abdominal pain) was present in 36% of Test and 28% of Control participants at baseline
(ns); the prevalence increased to 61% and 52%, respectively, at week 2 and fell to 47% and
42% by week 4 (differences between Test and Control were not significant). Furthermore,
the sum of the scores for the 5 major GI symptoms did not differ between Test and Control,
respectively, at any visit with mean
±
SD as follows: screen 2, 0.95
±
1.48 vs. 0.81
±
1.55;
baseline, 0.77
±
1.39 vs. 0.53
±
1.29; week 2, 1.67
±
2.12 vs. 1.45
±
2.09; week 4, 1.20
±
1.83
vs. 0.91 ±1.39.
3.2. Non-GI Symptoms
The number of subjects reporting symptom severity of none, mild, moderate or severe
for each visit on Test and Control are shown in Supplementary Tables S2 and S3. The
prevalence and severity of the 12 most common symptoms are shown in Figure 2.
Nutrients 2021,13, 1534 6 of 14
Nutrients 2021, 13, x FOR PEER REVIEW 6 of 14
3.2. Non-GI Symptoms
The number of subjects reporting symptom severity of none, mild, moderate or
severe for each visit on Test and Control are shown in Supplementary Tables S2 and S3.
The prevalence and severity of the 12 most common symptoms are shown in Figure 2.
Headache
0
10
20
30
40
50
60
70
A
Fatigue
*
B
Limb/Joint Pain
C
Anxiety
0
10
20
30
40
50
DLack of Energy
EExhaus tion
*
F
Reduced Ability
to Concentrate
0
10
20
30
40
50
Scr2 Base W k2 Wk4
JHot Flashes
Scr2 Base W k2 Wk4
KFeeling Cold
Scr2 Base W k2 Wk4
L
Lack of Appetite
0
10
20
30
40
50
GGloomy Thoughts
HInner Tension
I
Percentage of Participants with Symptom
% of Participant s
0
10
20
30
40
Wk2 Wk4
p=0.032
% of Participant s
0
10
20
30
40
Wk2 Wk4
*
% of Participant s
0
10
20
30
40
Wk2 Wk4
*
% of Participan ts
0
10
20
30
Wk2 Wk4
p=0.059
*
**
*
% of Participan ts
0
10
20
30
Wk2 Wk4
**
**
% of Participan ts
0
10
20
30
Wk2 Wk4
***
% of Participan ts
0
5
10
15
20
Wk2 Wk4
% of Participan ts
0
5
10
15
20
Wk2 Wk4
% of Participan ts
0
5
10
15
20
Wk2 Wk4
**
**
% of Participan ts
0
5
10
15
20
25
Wk2 Wk4
**
% of Participan ts
0
5
10
15
20
25
Wk2 Wk4
*
% of Participan ts
0
5
10
15
20
25
Wk2 Wk4
*
*
p=0.040
Figure 2. Prevalence and severity of headache (A), fatigue (B), limb/joint pain (C), anxiety (D), lack of energy (E),
exhaustion (F), lack of appetite (G), gloomy thoughts (H), inner tension (I), reduced ability to concentrate (J), hot flashes
(K) and feeling cold (L). Main panels: Values are means ± SD for n = 96 on Test (blue symbols and bars) and n = 95 on
Control (green symbols and bars). Scr2 = second screening visit; Base = baseline; Week2 = week 2, Wk4 = week 4. *
Prevalence significantly different from that at the Baseline visit by chi-squared test (p < 0.05). Insets: Percentage of
Figure 2.
Prevalence and severity of headache (
A
), fatigue (
B
), limb/joint pain (
C
), anxiety (
D
), lack of energy (
E
), exhaustion
(
F
), lack of appetite (
G
), gloomy thoughts (
H
), inner tension (
I
), reduced ability to concentrate (
J
), hot flashes (
K
) and
feeling cold (
L
). Main panels: Values are means
±
SD for n= 96 on Test (blue symbols and bars) and n= 95 on Control
(green symbols and bars). Scr2 = second screening visit; Base = baseline; Week2 = week 2, Wk4 = week 4. * Prevalence
significantly different from that at the Baseline visit by chi-squared test (p< 0.05). Insets: Percentage of participants in whom
the symptom was more severe (dark bars) or less severe (light bars) than the severity before treatment (median severity at
Scr2 and Base). The percent of participants with no change in severity is not shown. p-values are the significance of the
difference between the ratio of M:L on Test vs. Control (p< 0.05 by Fisher’s exact test). * Ratio of M:L significantly different
from 1:1 (p< 0.05 by binomial distribution) where M and L are the number of participants in whom the symptom was more
severe (M) or less severe (L).
Nutrients 2021,13, 1534 7 of 14
The presence or severity of 7 of the symptoms at Baseline did not differ signifi-
cantly by age, sex or BMI. The presence of Limb/Joint pain at Baseline differed by sex
(females > males) and age (over 48 years > under 48 years) and the severity of Limb/Joint
pain was greater in obese compared to lean or overweight subjects. The presence of fatigue
and hot flashes were affected by sex (females > males) and age (over 48 years > under
48 years). The severity of lack of energy was greater in females than males, whereas feeling
cold was more common in males than females (Supplementary Table S4).
The prevalence of symptoms did not differ significantly between Test and Control for
any symptom at any time. However, on Test the prevalence of fatigue was lower at week
4 and exhaustion at week 2 compared to baseline (Figure 2B,F). The change in severity
of symptoms did not differ significantly between Test and Control except for headache
and anxiety (less severe on Test vs. Control at week 2) and feeling cold at week 4 (insets
on Figure 2A,D,L). Compared to before treatment (median of scores at the Screen 2 and
Baseline visits), symptoms were less severe as follows: fatigue (Test at week 4), lack of
energy (both Test and Control at week 2 and week 4), exhaustion (Test at week 2 and week
4 and Control at week 4), limb/joint pain (Test at week 2), hot flashes (Test at week 4),
feeling cold (Control at week 2 and Test and week 4); anxiety and inner tension (Test and
Control at week 2 and week 4); and reduced ability to concentrate (Test at week 2 and week
4) (Figure 2). If the severity of symptoms at during treatment is compared to that at the
Baseline visit alone (Supplementary Table S5), fewer participants have more severe or less
severe symptoms, but the direction of the effects are similar to those shown in Figure 2.
The presence of symptoms at baseline was associated with increased serum CRP
(mg/L) (median [25%, 25%]) as follows (absence vs. presence, respectively): lack of
appetite, 1.31 [0.66, 3.36], n= 167 vs. 2.52 [1.48, 4.15] n= 24, p= 0.013; exhaustion, 1.30 [0.65,
3.40], n= 138 vs. 2.12 [1.07, 3.91], n= 53, p= 0.005; and limb/joint pain, 1.30 [0.65, 3.03],
n= 127 vs. 2.11 [1.08, 4.12], n= 64, p= 0.009. Although the presence of headache at baseline
was not associated with higher serum CRP, in participants whose CRP did not increase
during the study the severity of headache was greater on Control than Test (p= 0.049;
Figure 3A). The severity of exhaustion was greater in participants whose CRP increased vs.
those in whom it did not, however, changes in CRP did not affect the difference between
Test and Control (Figure 3B).
Serum oxLDL was not associated with the presence of any symptom at baseline. The
severity of fatigue was similar for participants in whom oxLDL increased during treatment
compared to those in whom it did not. However, in participants whose oxLDL did not
increase, fatigue was more severe on Control than Test (p= 0.052, Figure 3C). The severity
of hot flashes was greater in participants in whom oxLDL increased compared to those in
whom it did not (p= 0.049), but the severity of hot flashes on Test was similar to Control
regardless of the change in oxLDL (Figure 4D).
Nutrients 2021,13, 1534 8 of 14
Nutrients 2021, 13, x FOR PEER REVIEW 8 of 14
Headache
% of Participants
0
10
20
30
40
50
More Sever e
Less Severe
p=0.049
A
Exhaustio n
% of Partici pants
0
10
20
30
40
CRP CRP = =
CRP
p=0.045
B
**
*
Fatigue
% of Participants
0
10
20
30
40
50
Mor e Sever e
Less Severe
p=0.052
C
*
*
*
Hot Flashes
% of Participants
0
10
20
30
oxLDL oxLDL = =
oxLDL
p=0.049
D
*
*
Figure 3. Effect of changes in serum CRP and oxLDL on symptom severity. Percent of participants whose symptoms
became more severe (dark bars) or less severe (light bars) during treatment for those whose serum CRP (panels A and B)
or oxLDL (panels C and D) increased () or did not increase (=) after 4 weeks treatment. The percent of participants with
no change in severity is not shown. Brown/orange bars are for all participants, blue bars for Test and green bars for Control.
p-values over 2 bars are the significance of the difference between the ratios of More:Less severe for the 2 bars (eg. panel
A, in participants whose CRP did not increase after 4 weeks treatment, Headache was significantly more severe in those
on Control vs. Test p = 0.049). * Ratio of more:less severe differs significantly from 1:1 by chi-square test (p < 0.05).
Serum oxLDL was not associated with the presence of any symptom at baseline. The
severity of fatigue was similar for participants in whom oxLDL increased during
treatment compared to those in whom it did not. However, in participants whose oxLDL
did not increase, fatigue was more severe on Control than Test (p = 0.052, Figure 3C). The
severity of hot flashes was greater in participants in whom oxLDL increased compared to
those in whom it did not (p = 0.049), but the severity of hot flashes on Test was similar to
Control regardless of the change in oxLDL (Figure 4D).
At baseline, 11 of the 12 non-GI symptoms were significantly more common at in the
n = 88 participants with GI symptoms than in the n = 103 without GI symptoms (Table 1).
Additionally, changes in the severity of GI symptoms during the trial were associated
with differences in the severity of several non-GI symptoms. The overall severity of
headache was not associated with the severity of GI symptoms at either 2 weeks or 4
weeks. However, at 2 weeks, headache was less severe on Test vs. Control in participants
with no increase in the severity of GI symptoms (p = 0.021, Figure 4A). The overall severity
of fatigue at 2 weeks was lower in participants whose GI symptoms became more severe
compared to those in whom they did not (p = 0.027). The same was true for participants
on the Control treatment (p = 0.017). However, the severity of Fatigue on Test treatment
was not associated with differences in the severity of GI symptoms (Figure 4B). The
overall severity of joint/limb pain was greater at 2 weeks in participants with more severe
vs. the same or less severe GI symptoms (p = 0.046). The same was true for Control
participants at 2 weeks (p = 0.011) and 4 weeks (p = 0.049) but not for Test participants (ns).
At 2 weeks, the severity of joint/limb pain was lower on Test vs. Control in participants
with more severe GI symptoms than at baseline (p = 0.038, Figure 4C).
Figure 3.
Effect of changes in serum CRP and oxLDL on symptom severity. Percent of participants whose symptoms
became more severe (dark bars) or less severe (light bars) during treatment for those whose serum CRP (panels
A
and
B
) or
oxLDL (panels
C
and
D
) increased (
) or did not increase (=
) after 4 weeks treatment. The percent of participants with no
change in severity is not shown. Brown/orange bars are for all participants, blue bars for Test and green bars for Control.
p-values over 2 bars are the significance of the difference between the ratios of More:Less severe for the 2 bars (e.g., panel A,
in participants whose CRP did not increase after 4 weeks treatment, Headache was significantly more severe in those on
Control vs. Test p= 0.049). * Ratio of more:less severe differs significantly from 1:1 by chi-square test (p< 0.05).
Nutrients 2021, 13, x FOR PEER REVIEW 9 of 14
Figure 4. Effect of changes in GI symptoms on the severity of the non-GI symptoms headache (A), fatigue (B) and pains
in joints or limbs (C). Bars show the percent of participants whose non-GI symptoms became more severe (dark bars) or
less severe (light bars) during treatment for those whose GI symptoms (sum of scores for flatulence, diarrhea, constipation,
abdominal distention, and abdominal pain) became more severe (GI) or not (GI = ) during treatment. The percent of
participants with no change in severity is not shown. Brown bars show all participants, blue bars Test and green bars
Control. p-values are the significance of the difference between the ratios of More:Less severe for the bars indicated (e.g.,
panel A, in participants whose GI symptoms did not become more severe after 2 weeks treatment, Headache was more
severe on Control vs. Test p = 0.021). * Ratio of more:less severe differs significantly from 1:1 by chi-square test (p < 0.05).
Table 1. Association of non-GI symptoms with major GI symptoms.
Non-GI Symptom Major GI Symptom p
Absent (n = 103) Present (n = 88)
Headache 16 (16) 28 (32) 0.008
Fatigue 20 (19) 46 (52) <0.001
Pains in joints or limbs 8 (8) 35 (40) <0.001
Feelings of anxiety 12 (12) 27 (31) 0.001
Lack of energy 7 (7) 39 (44) <0.001
Tend to become exhausted 8 (8) 26 (30) <0.001
Lack of appetite 6 (6) 11 (13) 0.11
Gloomy thoughts 3 (3) 13 (15) 0.003
Inner tension 4 (4) 14 (16) 0.005
Diminished ability to concentrate 2 (2) 18 (20) <0.001
Hot flashes 10 (10) 25 (28) <0.001
Feeling cold 5 (5) 23 (26) <0.001
Values are number (%) of participants with each non-GI symptom at baseline amongst the
participants without any major GI symptom (flatulence, diarrhea, constipation, abdominal
distension and abdominal pain) compared to those with one or more major GI symptom at
screening or baseline. The right-most column shows the significance of the differences by chi-
squared test.
4. Discussion
The present results provide hypothesis-generating evidence that the OBG-enriched
oat product tested may influence several affective and physical feeling states in healthy
adults with LDL cholesterol between 3 and 5 mmol/L. Furthermore, markers of systemic
inflammation and oxidative stress and the severity of GI symptoms modified some of
these effects. These findings are consistent with current concepts about how alterations in
the gut-brain axis (increased colonic fermentation and alteration of the gut microbiota)
influence metabolism, behavior and brain function by a variety of different mechanisms
including effects on afferent pathways between the gut and the brain, short-chain fatty
acid (SCFA) production and other microbial by-products and metabolites which have
local and systemic effects on gut hormones, oxidative stress and inflammation [17,25].
Figure 4.
Effect of changes in GI symptoms on the severity of the non-GI symptoms headache (
A
), fatigue (
B
) and pains
in joints or limbs (
C
). Bars show the percent of participants whose non-GI symptoms became more severe (dark bars) or
less severe (light bars) during treatment for those whose GI symptoms (sum of scores for flatulence, diarrhea, constipation,
abdominal distention, and abdominal pain) became more severe (GI
) or not (GI =
) during treatment. The percent of
participants with no change in severity is not shown. Brown bars show all participants, blue bars Test and green bars Control.
p-values are the significance of the difference between the ratios of More:Less severe for the bars indicated (e.g., panel
A
,
in participants whose GI symptoms did not become more severe after 2 weeks treatment, Headache was more severe on
Control vs. Test p= 0.021). * Ratio of more:less severe differs significantly from 1:1 by chi-square test (p< 0.05).
At baseline, 11 of the 12 non-GI symptoms were significantly more common at in the
n= 88 participants with GI symptoms than in the n= 103 without GI symptoms (Table 1).
Additionally, changes in the severity of GI symptoms during the trial were associated with
differences in the severity of several non-GI symptoms. The overall severity of headache
was not associated with the severity of GI symptoms at either 2 weeks or 4 weeks. However,
at 2 weeks, headache was less severe on Test vs. Control in participants with no increase
in the severity of GI symptoms (p= 0.021, Figure 4A). The overall severity of fatigue at
Nutrients 2021,13, 1534 9 of 14
2 weeks was lower in participants whose GI symptoms became more severe compared
to those in whom they did not (p= 0.027). The same was true for participants on the
Control treatment (p= 0.017). However, the severity of Fatigue on Test treatment was not
associated with differences in the severity of GI symptoms (Figure 4B). The overall severity
of joint/limb pain was greater at 2 weeks in participants with more severe vs. the same
or less severe GI symptoms (p= 0.046). The same was true for Control participants at
2 weeks (p= 0.011) and 4 weeks (p= 0.049) but not for Test participants (ns). At 2 weeks,
the severity of joint/limb pain was lower on Test vs. Control in participants with more
severe GI symptoms than at baseline (p= 0.038, Figure 4C).
Table 1. Association of non-GI symptoms with major GI symptoms.
Non-GI Symptom Major GI Symptom p
Absent (n= 103) Present (n= 88)
Headache 16 (16) 28 (32) 0.008
Fatigue 20 (19) 46 (52) <0.001
Pains in joints or
limbs 8 (8) 35 (40) <0.001
Feelings of anxiety 12 (12) 27 (31) 0.001
Lack of energy 7 (7) 39 (44) <0.001
Tend to become
exhausted 8 (8) 26 (30) <0.001
Lack of appetite 6 (6) 11 (13) 0.11
Gloomy thoughts 3 (3) 13 (15) 0.003
Inner tension 4 (4) 14 (16) 0.005
Diminished ability to
concentrate 2 (2) 18 (20) <0.001
Hot flashes 10 (10) 25 (28) <0.001
Feeling cold 5 (5) 23 (26) <0.001
Values are number (%) of participants with each non-GI symptom at baseline amongst the participants without any
major GI symptom (flatulence, diarrhea, constipation, abdominal distension and abdominal pain) compared to
those with one or more major GI symptom at screening or baseline. The right-most column shows the significance
of the differences by chi-squared test.
4. Discussion
The present results provide hypothesis-generating evidence that the OBG-enriched
oat product tested may influence several affective and physical feeling states in healthy
adults with LDL cholesterol between 3 and 5 mmol/L. Furthermore, markers of systemic
inflammation and oxidative stress and the severity of GI symptoms modified some of
these effects. These findings are consistent with current concepts about how alterations
in the gut-brain axis (increased colonic fermentation and alteration of the gut microbiota)
influence metabolism, behavior and brain function by a variety of different mechanisms
including effects on afferent pathways between the gut and the brain, short-chain fatty acid
(SCFA) production and other microbial by-products and metabolites which have local and
systemic effects on gut hormones, oxidative stress and inflammation [17,25].
The association between the presence of GI symptoms and the increased prevalence of
11 of the 12 non-GI symptoms is not a new finding and could be ascribed to psychological
or physiological factors. For example, it is known that the perception of GI symptoms
such as constipation and flatulence are not related to the amount of gas in the colon [
26
]
and are associated with personality and anxiety [
27
]. Thus, participants who noticed
GI symptoms may have been, in general, more aware of other abnormal sensations and
feelings they experienced and were more able to remember to report them. However, the
association between GI and non-GI symptoms could have a physiological basis via the
gut-brain axis. There is abundant evidence from studies
in vitro
and in animal models
that oats and OBG influence SCFA production and the colonic microbiome [
14
], although
the results are inconsistent and there are few studies in humans. OBG-enriched oat bran
containing 20 g/d dietary fiber and 10 g/d OBG did not increase fecal SCFA after 4 weeks
Nutrients 2021,13, 1534 10 of 14
but did so at 8 and 12 weeks in healthy subjects [
28
]. Furthermore, fecal SCFA were not
increased by either granola containing 6 g/d fiber and 3 g/d OBG daily for 5 weeks [
29
] or
by 60 g oatmeal containing 8.5 g dietary fiber and 4.7 g OBG daily for 1 week [
30
]. However,
the granola increased fecal bifidobacteria and lactobacilli [
29
] and the oatmeal influenced
bacterial metabolism as judged by reduce
β
-galactosidase and urease concentrations [
30
].
A recent study showed, in hypercholesterolemic subjects that compared to 80 g white
rice, 80 g oatmeal daily for 45 days increased the abundance of fecal Firmicutes [
31
]. The
effect of OBG on the fecal microbiome might also by its molecular weight, since 3 g high
molecular weight (1349 kDa) barley
β
-glucan for 5 weeks significantly increased fecal
Bacteriodetes and reduced Firmicutes compared to a
β
-glucan-free control and to 3 g low
molecular weight (288 kDa) barley
β
-glucan [
32
]. Thus, there is evidence in humans that
the amount of OBG we provided could influence the fecal microbiome, but not fecal SCFA.
However, the composition of the fecal microbiome may not reflect that of the mucosa-
associated micro-organisms [
33
]. In addition, there is evidence in humans that increased
SCFA production can occur in the absence of a change in fecal SCFA [
34
] and that a high
fecal acetate concentration may reflect reduced acetate absorption rather than increased
production [35].
The severity of headache was lower on Test than Control at 2 weeks in all subjects
(Figure 2A) and at 4 weeks in those in whom serum CRP did not increase (Figure 3A).
The causes of headaches are not well understood, but may include hypoglycemia [
36
],
obesity [
37
] or inflammation [
25
]. However, the n= 67 participants with headache at
baseline had similar (mean
±
SD) BMI, 28.08
±
4.70 kg/m
2
, and median CRP, 1.52 mg/L,
as the n= 127 without headache, 27.89 ±4.58 kg/m2, and 1.53 mg/L.
Fatigue became less common and less severe than at baseline after 4 weeks treatment with
OBG, as did exhaustion after 2 weeks, but the differences from control were not significant.
Lack of energy was less severe on both Test and Control treatments compared to baseline.
These results are in line with studies showing that OBG [
21
] and konjac oligosaccharide [
38
]
increased maximum exercise time in rats or mice and altered metabolites suggestive of
reduced fatigue. In humans, consuming cereals high in wheat-fiber for 2 weeks reduced
fatigue compared to control in healthy adults [
19
] and 13 weeks treatment with a prebiotic
containing inulin and fructo-oligosaccharides reduced exhaustion in elderly subjects when
compared to control [
39
]. Fatigue is associated with lack of sleep, an association that may be
mediated by increased inflammation [
40
]. At baseline, serum CRP was similar in participants
with and without fatigue (not shown) and with or without lack of energy (not shown), but
the presence of exhaustion at baseline was associated with a higher CRP. Furthermore, the
severity of exhaustion increased from baseline to a greater extent in participants whose CRP
increased compared to those in whom it did not (Figure 3B).
Pains in muscles can be due to overuse or to reduced circulation, while pain in joints
is most commonly due to intervertebral disc herniation or osteoarthritis, both of which
are associated with obesity, chronic inflammation [
41
43
] and older age. This is consistent
with our data in that the n= 64 participants with limb/joint pain at baseline were older
(mean
±
SD, 52.1
±
10.3 vs. 45.3
±
11.3 years, p< 0.001), and had higher BMI (29.1
±
4.5
vs. 27.3
±
4.6 kg/m
2
,p= 0.013), and higher CRP (2.11 [1.08, 4.12] vs. 1.30 [0.65, 3.03] mg/L,
p= 0.009) than the n= 127 without limb/joint pain. After 2 weeks treatment, limb/joint
pain was less severe than at baseline in all subjects on OBG (Figure 2C), and, in participants
with more severe GI symptoms, less severe on OGB compared to control at 2 weeks
(Figure 4C). On Control, but not OBG, having more severe GI symptoms was associated
with more severe limb/joint pain at 2 and 4 weeks (Figure 4C). These observations, along
with the increased prevalence of limb/joint pain in those with GI symptoms (Table 1)
suggest a link between the gut and joint/limb pain. There is some evidence that people with
rheumatoid arthritis (RA) have different colonic microbiota and that probiotic treatment
can improve the condition in the absence of changes in CRP [
44
] but this may not apply to
our study population, most of whom likely did not have RA.
Nutrients 2021,13, 1534 11 of 14
During the study, the severity of anxiety decreased from baseline on both treatments,
with the reduction on OBG being greater than that on Control at 2 weeks (p= 0.059, Figure 2D).
A link between the colonic microbiota and anxiety is suggested by a study in which a probiotic
formulation containing Lactobacillus helveticus R0052 and Bifidobacterium longum R0175, given
to male Wistar rats for 2 weeks, reduced anxiety-like behavior, and given to humans for
30 d reduced psychological distress, the Hospital Anxiety and Depression Scale and urinary
free-cortisol excretion [
45
]. Additionally, compared to control, 5.5 g/d galactooligosaccharides
for 3 weeks, reduced the salivary cortisol waking response and increased the processing of
positive vs. negative attentional vigilance in healthy subjects, although these effects were not
seen with fructooligosaccharides [46].
The prevalence of feeling cold at 4 weeks did not change from baseline on either
treatment, but the severity of feeling cold at 4 weeks was less on Test than Control (Figure 2L).
Consistent with several studies in which feeling cold was associated with musculoskeletal
pain [
47
49
], we found that feeling cold was present in 41.9% of 43 participants who had
joint/limb pain at baseline but only 6.8% of 148 participants who did not (p< 0.001).
Furthermore, after 4 weeks, the severity of feeling cold had increased from baseline in 20%
of the 20 participants who experienced more severe joint/limb pain compared to only 2.3%
of the 171 in who did not (p< 0.001). However, it is not clear why the prevalence and
severity of feeling cold would be less after 4 weeks on OBG vs. Control.
Hot flashes are a common symptom in post-menopausal women. In this study, 93.5%
of the 46 participants with hot flashes at baseline were women (p< 0.001). Furthermore,
although the mean
±
SD age of women with and without hot flashes at baseline were
similar, 52.5
±
9.8 vs. 49.6
±
11.4 year (p= 0.16), a higher percentage of n= 76 women
aged > 48 year had hot flashes compared to the n= 43 aged
48 year, 46 vs. 19%, p= 0.003.
Increased oxidative stress is associated with a greater severity of hot flashes [
50
], but
it is not clear if menopausal symptoms are a cause or an effect of oxidative stress [
51
].
In this study the concentration of oxLDL at baseline (median {95%CI}) did not differ in
the 46 participants with hot flashes at baseline, 63 {37, 124}
µ
g/L, compared to the 145
without, 78 {45, 144}
µ
g/L. However, it is of interest that the severity of hot flashes at week
4 compared to baseline increased more in participants in whom oxLDL increased compared
to those in whom it did not (Figure 3D).
The major weakness of this assessment of non-GI symptoms is that the study was
designed to assess the effect of OBG on serum cholesterol; assessment of symptoms was a
tertiary objective not listed in the study registration. Several hundred statistical analyses
were performed post hoc with no correction for multiple comparisons; this increases the
likelihood of making type 1 errors. Furthermore, validated questionnaires were not used to
assess the symptoms. Nevertheless, there was significant amount of internal and external
consistency of the results with what might be expected from the literature.
5. Conclusions
These results provide hypothesis-generating evidence that OBG may have a beneficial
effect on several affective and physical feeling states in healthy adults. Since there is a
paucity of information about the effects of dietary fiber in general, and oats and OBG
specifically, on non-GI symptoms in humans, these results provide information which may
be useful for designing the studies which would be required to confirm these observations.
Supplementary Materials:
The following are available online at https://www.mdpi.com/article/10
.3390/nu13051534/s1, Inclusion/exclusion criteria; Table S1: Nutritional composition of intervention
products (per sachet); Tables S2 and S3: Occurrence of non-GI symptoms on the symptoms question-
naire; Table S4: Effect of age, sex and BMI on symptoms at Baseline; Table S5: Severity of symptoms
when compared to severity at the Baseline visit.
Author Contributions:
Conceptualization, T.M.S.W., M.R. and Y.C.; methodology, T.M.S.W.; formal
analysis, T.M.S.W.; investigation, A.L.J., A.E. and J.E.C.; resources, M.R., E.H.D. and Y.C.; data
curation, A.E. and J.E.C.; writing—original draft preparation, T.M.S.W.; writing—review and editing,
Nutrients 2021,13, 1534 12 of 14
M.R., E.H.D., A.L.J., A.E., J.E.C. and Y.C.; visualization, T.M.S.W.; supervision, T.M.S.W.; project
administration, A.L.J., A.E. and J.E.C. All authors have read and agreed to the published version of
the manuscript.
Funding: This research was funded by PepsiCo.
Institutional Review Board Statement:
The study was conducted according to the guidelines of the
Declaration of Helsinki, and approved by the WIRB®(protocol #20190694, 28 March 2019).
Informed Consent Statement:
Informed consent was obtained from all subjects involved in the study.
Data Availability Statement:
The data presented in this study are available upon request from the
corresponding author.
Acknowledgments:
The authors wish to thank Kirstin Harris (PepsiCo) and Alison Kamil (PepsiCo)
for support in the early stages of the project.
Conflicts of Interest:
T.M.S.W. and A.L.J. are employees and part owners of INQUIS; A.E. and J.E.C.
are employees of INQUIS; M.R. was an employee at PepsiCo during the clinical and data analysis
phase; E.D. and Y.C. are employees of PepsiCo. INQUIS, T.M.S.W., A.E., J.E.C. and A.L.J. do not own
any intellectual property which may arise from this study. T.M.S.W., A.E., J.E.C. and A.L.J. have not
received any payments directly from PepsiCo and have no financial interest in PepsiCo. The funder
assisted in designing the study but had no role in the collection or analysis of the data. The funder
decided to publish these results and commented on the manuscript drafts; however, T.M.S.W. had
sole responsibility for determining its final content.
Disclaimer:
The views expressed in this manuscript are those of the authors and do not necessarily
reflect the position or policy of PepsiCo, Inc.
References
1.
Healthy Diet; Fact Sheet No. 394, Updated August 2018. World Health Organization. Available online: http://www.who.int/
mediacentre/factsheets/fs394?en/ (accessed on 20 November 2020).
2.
Wood, P.J. Oat
β
-glucan: Structure, location and properties. In Oats: Chemistry and Technology; Webster, F.H., Ed.; AACC Inc.:
St. Paul, MN, USA, 1986; pp. 121–152.
3.
Whitehead, A.; Beck, E.J.; Tosh, S.; Wolever, T.M.S. Cholesterol-lowering effects of oat
β
-glucan: A meta-analysis of randomized
controlled trials. Am. J. Clin. Nutr. 2014,100, 1413–1421. [CrossRef]
4.
Ho, H.V.T.; Sievenpiper, J.L.; Zurbau, A.; Mejia, S.B.; Jovanovski, E.; Au-Yeung, F.; Jenkins, A.L.; Vuksan, V. The effect of oat
β
-glucan on LDL-cholesterol, non-HDL-cholesterol and apoB for CVD risk reduction: A systematic review and meta-analysis of
randomized-controlled trials. Br. J. Nutr. 2016,116, 1369–1382. [CrossRef] [PubMed]
5.
Zurbau, A.; Noronha, J.; Khan, T.A.; Sievenpiper, J.L.; Wolever, T.M.S. The effect of oat
β
-glucan on postprandial blood glucose
and insulin responses: A systematic review and meta-analysis. Eur. J. Clin. Nutr.
2021
. published on line before print. [CrossRef]
[PubMed]
6.
Delahoy, P.J.; Magliano, D.J.; Webb, K.; Grobler, M.; Liew, D. The relationship between reduction in low-density lipoprotein
cholesterol by statins and reduction in risk of cardiovascular outcomes: An updated metaanalysis. Clin. Ther.
2009
,31, 236–244.
[CrossRef] [PubMed]
7.
Livesey, G.; Taylor, R.; Livesey, H.F.; Buyken, A.E.; Jenkins, D.J.A.; Augustin, L.S.A.; Sievenpiper, J.L.; Barclay, A.W.; Liu, S.;
Wolever, T.M.S.; et al. Dietary glycemic index and load and the risk of type 2 diabetes: A systematic review and updated
meta-analysis of prospective cohort studies. Nutrients 2019,11, 1280. [CrossRef]
8.
Grabistske, H.A.; Slavin, J.L. Gastrointestinal effects of low-digestible carbohydrates. Crit. Rev. Food Sci. Nutr.
2009
,49, 327–360.
[CrossRef]
9.
Wolever, T.M.S.; Rahn, M.; Dioum, E.; Spruill, S.E.; Ezatagha, A.; Campbell, J.E.; Jenkins, A.L.; Chu, Y. Impact of an oat
β
-glucan
beverage on LDL-cholesterol and cardiovascular risk in men and women with borderline high cholesterol: A double-blind,
randomized, controlled clinical-trial. J. Nutr. 2021, in press.
10.
Kaukinen, K.; Collin, P.; Huhtala, H.; Mäki, M. Long-term consumption of oats in adult celiac disease patients. Nutrients
2013
,5,
4380–4389. [CrossRef]
11.
Ford, A.C.; Moayyedi, P.; Lacy, B.E.; Lembo, A.J.; Saito, Y.A.; Schiller, L.R.; Soffer, E.E.; Spiegel, B.M.; Quigley, E.M. American
College of Gastroenterology monograph on the management of irritable bowel syndrome and chronic idiopathic constipation.
Am. J. Gastroenterol. 2014,109 (Suppl. 1), S2–S26. [CrossRef] [PubMed]
12. Humes, D.; Smith, J.K.; Spiller, R.C. Colonic diverticular disease. BMJ Clin. Evid. 2011,2011, 0405.
13.
Cumming, J.H. The effect of dietary fiber on fecal weight and composition. In CRC Handbook of Dietary Fiber in Human Nutrition,
3rd ed.; Spiller, G.A., Ed.; CRC Press: Roca Raton, FL, USA, 1985; pp. 183–252.
Nutrients 2021,13, 1534 13 of 14
14.
Korczak, R.; Kocher, M.; Swanson, K.S. Effects of oats on gastrointestinal health as assessed by
in vitro
, animal and human studies.
Nutr. Rev. 2020,78, 343–363. [CrossRef]
15. Swann, O.G.; Kilpatrick, M.; Breslin, M.; Oddy, W.H. Dietary fiber and its associations with depression and inflammation. Nutr.
Rev. 2020,78, 394–411. [CrossRef]
16.
Luca, M.; Di Mauro, M.; Di Maruo, M.; Luca, A. Gut microbiota in Alzheimer’s disease, depression, and type 2 diabetes mellitus:
The role of oxidative stress. Oxid. Med. Cell Longev. 2019,2019, 4730539. [CrossRef] [PubMed]
17.
Morais, L.H.; Schrieber, H.L.; Mazmanian, S.K. The gut microbiota-brain axis in behaviour and brain disorders. Nat. Rev. Microbiol.
2021,19, 241–255. [CrossRef]
18.
O’Reilly, G.A.; Huh, J.; Schembre, S.M.; Tate, E.B.; Pentz, M.A.; Dunton, G. Association of usual self-reported dietary intake with
ecological momentary measures of affective and physical feeling states in children. Appetite 2015,92, 314–321. [CrossRef]
19.
Smith, A.; Bazzoni, C.; Beale, J.; Elliott-Smith, J.; Tiley, M. High fibre breakfast cereals reduce fatigue. Appetite
2001
,37, 249–250.
[CrossRef]
20.
Wolever, T.M.S.; Chiasson, J.-L.; Josse, R.G.; Leiter, L.A.; Maheux, P.; Rabasa-Lhoret, R.; Rodger, N.W.; Ryan, E.A. Effects of
changing the amount and source of dietary carbohydrates on symptoms and dietary satisfaction over a 1-year period in subjects
with type 2 diabetes: Canadian trial of carbohydrates in diabetes (CCD). Can. J. Diab. 2017,41, 164–176. [CrossRef] [PubMed]
21.
Xu, C.; Lv, J.; Lo, M.; Cui, S.W.; Hu, X.; Fan, M. Effects of oat
β
-glucan on endurance exercise and its anti-fatigue properties in
trained rats. Carbohydr. Polym. 2013,92, 1159–1165. [CrossRef]
22.
Chiasson, J.-L.; Josse, R.G.; Hunt, J.A.; Palmason, C.; Rodger, N.W.; Ross, S.A.; Ryan, E.A.; Tan, M.H.; Wolever, T.M.S. The efficacy
of acarbose in the treatment of patients with non-insulin-dependent diabetes mellitus. A multicenter controlled clinical trial. Ann.
Int. Med. 1994,121, 928–935. [CrossRef] [PubMed]
23.
Wolever, T.M.S.; Chiasson, J.-L.; Josse, R.G.; Hunt, J.A.; Palmason, C.; Rodger, N.W.; Ross, S.A.; Ryan, E.A.; Tan, M.H. No
relationship between carbohydrate intake and effect of acarbose on HbA1c or gastrointestinal symptoms in type 2 diabetic
subjects consuming 30–60% of energy from carbohydrate. Diabetes Care 1998,21, 1612–1618. [CrossRef]
24.
Wolever, T.M.S.; Tosh, S.M.; Gibbs, A.L.; Brand-Miller, J.; Duncan, A.M.; Hart, V.; Lamarche, B.; Thomson, B.A.; Duss, R.; Wood,
P.J. Physicochemical properties of oat
β
-glucan influence its ability to reduce serum LDL cholesterol in humans: A randomized
clinical trial. Am. J. Clin. Nutr. 2010,92, 723–732. [CrossRef]
25. Hindiyeh, N.; Aurora, S.K. What the gut can teach us about migrane. Curr. Pain Headache Rep. 2015,19, 33. [CrossRef]
26.
Lasser, R.B.; Bond, J.H.; Levitt, M.D. The role of intestinal gas in functional abdominal pain. N. Eng. J. Med.
1975
,293, 524–526.
[CrossRef] [PubMed]
27.
Devroede, G.; Girard, G.; Bouchoucha, M.; Roy, T.; Black, R.; Camerlain, M.; Pinard, G.; Schang, J.-C.; Arhan, P. Idiopathic
constipation by colonic dysfunction: Relationship with personality and anxiety. Dig. Dis. Sci.
1989
,34, 1428–1433. [CrossRef]
[PubMed]
28.
Nilsson, U.; Johansson, M.; Nilsson, A.; Björk, I.; Nyman, M. Dietary supplementation with
β
-glucan enriched oat bran increases
faecal concentration of carboxylic acids in healthy subjects. Eur. J. Clin. Nutr. 2008,62, 978–984. [CrossRef] [PubMed]
29.
Connolly, M.L.; Tzounis, X.; Tuohy, K.M.; Lovegrove, J.A. Hypocholesterolemic and probiotic effects of a whole-grain oat-based
granola breakfast cereal in a cardio-metabolic “at risk” population. Front. Microbiol. 2016,7, 1675. [CrossRef]
30.
Valeur, J.; Puaschitz, N.G.; Midtvedt, T.; Berstad, A. Oatmeal porridge: Impact on microflora-associated characteristics in healthy
subjects. Brit. J. Nutr. 2016,115, 62–67. [CrossRef] [PubMed]
31.
Ye, M.; Sun, J.; Chen, Y.; Ren, Q.; Li, Z.; Zhao, Y.; Pan, Y.; Xue, H. Oatmeal induced gut microbiota alteration and its relationship
with improved lipid profiles: A secondary analysis of a randomized clinical trial. Nutr. Metab. (Lond.)
2020
,17, 85. [CrossRef]
[PubMed]
32.
Wang, Y.; Ames, N.P.; Tun, H.M.; Tosh, S.M.; Jones, P.J.; Khafipour, E. High molecular weight barley
β
-glucan alters gut microbiota
toward reduced cardiovascular disease risk. Front. Micriobiol. 2016,7, 129. [CrossRef]
33.
Zoetendal, E.G.; von Wright, A.; Vilpponen-Salmela, T.; Ban-Amor, K.; Akkermans, A.D.L.; de Vos, W.M. Mucosa-associated
bacteria in the human gastrointestinal tract are uniformly distributed along the colon and different from the community recovered
from feces. Appl. Environ. Microbiol. 2002,68, 3401–3407. [CrossRef] [PubMed]
34.
Vogt, J.A.; Ishii-Schrade, K.B.; Pencharz, P.B.; Wolever, T.M.S. L-Ramnose increases serum propionate after long-term supplemen-
tation, but lactulose does not raise serum acetate. Am. J. Clin. Nutr. 2004,80, 1254–1261. [CrossRef] [PubMed]
35.
Vogt, J.A.; Wolever, T.M.S. Fecal acetate is inversely related to acetate absorption from the human rectum and distal colon. J. Nutr.
2003,133, 3145–3148. [CrossRef]
36. Torelli, P.; Manzoni, G.C. Fasting headache. Curr. Pain Headache Rep. 2010,14, 284–291. [CrossRef] [PubMed]
37.
Bigal, M.E.; Rapoport, A.M. Obesity and chronic daily headache. Curr. Pain Headache Rep.
2010
,16, 101–109. [CrossRef] [PubMed]
38.
Zeng, Y.; Zhang, J.; Zhang, Y.; Men, Y.; Zhang, B.; Sun, Y. Prebiotic, immunomodulating and anti-fatigue effects of konjac
oligosaccharide. J. Food Sci. 2018,83, 3110–3117. [CrossRef]
39.
Buigues, C.; Fernández-Garrido, J.; Pruimboom, L.; Hoogland, A.J.; Navarro-Martínez, R.; Martínez-Martínez, M.; Verdejo, Y.;
Mascarós, M.C.; Peris, C.; Cauli, O. Effect of a prebiotic formulation on frailty syndrome: A randomized, double-blind clinical
trial. Int. J. Mol. Sci. 2016,17, 932. [CrossRef]
40.
Thomas, K.S.; Motivala, S.; Olmstead, R.; Irwin, M.R. Sleep depth and fatigue: Role of cellular inflammatory activation. Brain
Behav. Immun. 2011,25, 53–58. [CrossRef]
Nutrients 2021,13, 1534 14 of 14
41.
Lean, M.E.J.; Han, T.S.; Seidell, J.C. Impairment of health and quality of life in people with large waist circumference. Lancet
1998
,
351, 853–856. [CrossRef]
42. Schaible, H.G. Spinal mechanisms contributing to joint pain. Novartis Found. Symp. 2004,260, 4–22.
43.
Sanghi, D.; Mishra, A.; Sharma, A.C.; Raj, S.; Mishra, R.; Natu, S.M.; Agarwal, S.; Srivastava, R.N. Elucidation of dietary risk
factors in osteoarthritis knee: A case-control study. J. Am. Coll. Nutr. 2015,34, 15–20. [CrossRef]
44.
Bravo-Blas, A.; Wessel, H.; Milling, S. Microbiota and arthritis: Correlations or cause? Curr. Opin. Rheumatol.
2016
,28, 161–167.
[CrossRef] [PubMed]
45.
Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Najdi, A.; Bisson, J.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al.
Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium
longum R0175) in rats and human subjects. Brit. J. Nutr. 2011,105, 755–764. [CrossRef] [PubMed]
46.
Schmidt, K.; Cowen, P.J.; Harmer, C.H.; Tzortzis, G.; Errington, S.; Burnet, P.W.J. Prebiotic intake reduces the waking cortisol
response and alters emotional bias in healthy volunteers. Psychoparmacology 2015,232, 1793–1801. [CrossRef] [PubMed]
47.
Bang, B.E.; Aasmoe, L.; Aardal, L.; Andorsen, G.S.; Bjørnbakk, A.K.; Egeness, C.; Espejord, I.; Kramvik, E. Feeling cold at work
increases the risk of symptoms from muscles, skin and airways in seafood industry workers. Am. J. Ind. Med.
2005
,47, 65–71.
[CrossRef] [PubMed]
48.
Sormunen, E.; Remes, J.; Hassi, J.; Pienimäki, T.; Rintamäki, H. Factors associated with self-estimated work ability and mus-
culoskeletal symptoms among male and female workers in cooled food-processing facilities. Ind. Health
2009
,47, 271–282.
[CrossRef]
49.
Farbu, E.H.; Skandfer, M.; Nielsen, C.; Brenn, T.; Stubhaug, A.; Höper, A.C. Working in a cold environment, feeling cold at work
and chronic pain: A cross-sectional analysis of the Tromsø study. BMJ Open 2019,9, e031248. [CrossRef]
50.
Sánchez-Rodríguez, M.A.; Zacarías-Flores, M.; Arronte-Rosales, A.; Mendoza-Núñez, V.M. Association between hot flashes
severity and oxidative stress among Mexican postmenopausal women: A cross-sectional study. PLoS ONE
2019
,14, e0214264.
[CrossRef] [PubMed]
51.
Cervellati, C.; Bergamini, C.M. Oxidative damage and the pathogenesis of menopause related disturbances and diseases. Clin.
Chem. Lab. Med. 2016,54, 739–753. [CrossRef]
... Similarly, in healthy subjects experiencing fatigue in daily life, the consumption of 350 mg of euglenaderived β-glucan per day for four weeks led to a significant reduction in both physical and mental fatigue compared to the placebo group, although no significant difference was observed in overall fatigue [9]. Conversely, in healthy subjects with LDLcholesterol levels ranging from 3 to 5 mmol/L, the administration of 3000 mg of oat-derived β-glucan per day for four weeks did not result in a significant difference in the prevalence and severity of fatigue compared to the placebo group [10]. Thus, while various clinical trials have been conducted on the effects of β-glucans on fatigue, the results have shown inconsistency. ...
... Although the target population across studies was generally healthy, specific characteristics varied. For instance, one study focused on individuals with qi deficiency [22], another on those with LDL-cholesterol levels between 3 and 5 mmol/L [10], and additional studies targeted individuals with decreased vitality, sleep quality [23], feelings of fatigue [9], engagement in aerobic or endurance exercise [24,25], recurrent upper respiratory tract infections [26], marathon runners [8,27], allergies such as ragweed or hay fever [28], and varying levels of psychological stress [29,30]. Furthermore, one study focused on individuals with a history of colds [31]. ...
... Wolever [10] Fatigue: ...
Article
Full-text available
Several clinical trials suggest that β-glucans may reduce feelings of fatigue, however the results of clinical trials are inconsistent. Additionally, no systematic reviews or meta-analyses have assessed the effects of β-glucans on fatigue. Therefore, this study investigates the effects of β-glucans on fatigue in healthy subjects through a systematic review and meta-analysis. PubMed, Web of Science, and the Cochrane Central Register of Controlled Trials (CENTRAL) were searched from database inception to March 15, 2024. The inclusion criterion was a randomized controlled trial (RCT) investigating the effects of β-glucans on healthy subjects’ fatigue, vigor, and mood state. To assess risk of bias, we employed the Cochrane risk of bias tool. A random-effects meta-analysis was conducted for the standardized mean difference (SMD). Sixteen RCTs with a total of 1,449 participants were included, and 12 studies provided data suitable for meta-analysis. Meta-analysis revealed that β-glucans significantly reduced feelings of fatigue (SMD = −0.32, 95% CI = −0.53 to −0.12; p = 0.0021), increased vigor (SMD = 0.46, 95% CI = 0.26–0.66; p < 0.0001), and improved mood state (SMD = 0.32, 95% CI = 0.11–0.53; p = 0.0026) compared to the placebo group. The results of the systematic review and meta-analysis indicate that β-glucans may be effective in reducing feelings of fatigue in healthy individuals. However, the number of studies included is insufficient, suggesting that further clinical trials are needed to validate this effect.
... β-glucan regulates the gastrointestinal transit rate after meals and starch digestion to dilute blood sugar level through changes in the glycemic index, thereby reducing the insulinemic response in diabetes [41,45]. In addition, β-glucan has various physiological effects such as preventing cardiovascular diseases by controlling the blood pressure and anti-obesity and anti-cancer effects (e.g., prevention of colorectal cancer) [18,[46][47][48][49][50][51][52][53][54][55]. It lowers low-density lipoprotein (LDL) cholesterol level in the blood and elevates high-density lipoprotein (HDL) cholesterol level to help maintain normal blood lipid concentration and body weight [56]. ...
... However, some moderate adverse effects have been reported. Symptoms including ulcerative colitis, diarrhea, back pain, joint pain, kidney disease, circadian disruption-induced metabolic syndrome, bile acid storage and vascular calcium storage are reported to be ameliorated in the specific conditions such as ingestion with a high-doselimiting concentration of β-glucan [48,49,[65][66][67][68]. It remains to be elucidated whether this intake is safe for women who are pregnant or breastfeeding. ...
... In addition, dietary intake of β-glucan may be unsafe for an individual with certain disease-related conditions such as acquired immune deficiency syndrome (AIDS) and AIDS-related complex [69]. However, no serious adverse effects related to β-glucan supplementations have been reported during laboratory and clinical trials except in some special cases as mentioned above [49,70,71]. effects (e.g., prevention of colorectal cancer) [18,[46][47][48][49][50][51][52][53][54][55]. ...
Article
Full-text available
Oats (Avena sativa L.) are rich in protein, fiber, calcium, vitamins (B, C, E, and K), amino acids, and antioxidants (beta-carotene, polyphenols, chlorophyll, and flavonoids). β-glucan and avenanthramides improve the immune system, eliminate harmful substances from the body, reduce blood cholesterol, and help with dietary weight loss by enhancing the lipid profile and breaking down fat in the body. β-glucan regulates insulin secretion, preventing diabetes. Progladins also lower cholesterol levels, suppress the accumulation of triglycerides, reduce blood sugar levels, suppress inflammation, and improve skin health. Saponin-based avanacosidase and functional substances of flavone glycoside improve the immune function, control inflammation, and prevent infiltration in the skin. Moreover, lignin and phytoestrogen prevent hormone-related cancer and improve the quality of life of postmenopausal women. Sprouted oats are rich in saponarin in detoxifying the liver. The literatures have been reviewed and the recent concepts and prospects have been summarized with figures and tables. This review discusses recent trends in research on the functionality of oats rather than their nutritional value with individual immunity for self-medication. The oat and its acting components have been revisited for the future prospect and development of human healthy and functional sources.
... Interestingly, individuals who received beta-glucan had less severe headaches and lower perception of cold than controls. In addition, changes in inflammatory markers, C-reactiveprotein (CRP), and oxidized-LDL with gastrointestinal symptom severity were associated with the occurrence and severity of several non-GI symptoms [51]. Furthermore, the results of a phase I/II clinical trial conducted in patients with advanced malignancies receiving chemotherapy indicated that beta-(1,3)-(1,6)-D-glucan administered as an adjunctive therapy may improve white blood cells and platelet counts, and may also raise levels of hemoglobin, which in up to 40% of patients reduces the feeling of fatigue compared with the period before the supplementation [52]. ...
... Another preliminary study found that beta-glucan administration improved mood and vitality among HR-QoL measures in ME/CFS [51]. Overall, while there are some preliminary research studies suggesting possible potential benefits of beta-glucan administration on fatigue, mental health, and quality of life in ME/CFS, the current evidence is limited, and more research is needed to fully evaluate its efficacy and understand the pathophysiological mechanisms after beta-glucan administration in ME/CFS and other chronic post-infectious conditions. ...
Article
Full-text available
This research aimed to examine the potential alleviative effects of beta-glucan administration on fatigue, unrefreshing sleep, anxiety/depression symptoms and health-related quality of life in ME/CFS. A 36-week unicenter, randomized, double-blind, placebo-controlled trial was conducted in 65 ME/CFS patients, who were randomly allocated to one of two arms to receive four capsules each one of 250 mg beta-glucan, 3.75 µg vitamin D3, 1.05 mg vitamin B6, and 7.5 mg zinc (n = 35), or matching placebo including only microcrystalline cellulose as an excipient (n = 30) once daily. The findings showed that the beta-glucan supplementation significantly improved cognitive fatigue (assessed with FIS-40 scores) after the 36-week treatment compared to the baseline (p = 0.0338). Taken together, this study presents the novel finding that yeast-derived beta-glucan may alleviate cognitive fatigue symptoms in ME/CFS. Thus, it offers valuable scientific insights into the potential use of yeast beta-glucan as a nutritional supplement and/or functional food to prevent or reduce cognitive dysfunction in patients with ME/CFS. Further interventions are warranted to validate these findings and also to delve deeper into the possible immunometabolic pathomechanisms of beta-glucans in ME/CFS.
... For instance, common oat (Avena sativa) and its constituents have gained considerable attention not only as important food supplements but also as immunomodulators with potential effects on the prevention of age-related disease [31][32][33][34][35][36]. Salient compounds found in oats that may mediate its clinical benefit include -glucans, as demonstrated by multiple studies in humans where supplementation with -glucan extract resulted in important health benefits, including a significant reduction in total cholesterol and low-density lipoprotein and CVD risk [37][38][39][40][41]. Furthermore, the US database ClinicalTrials.gov ...
... Here, we conducted secondary measurements using serum samples from a doubleblinded placebo-controlled clinical study to investigate the effects of the consumption of an oat product (SoluOBC) providing 3 g of β-Glucan (OBG) compared to a rice control (PCB) after 2-and 4-weeks intervention (Supplementary Materials Figure S1) on blood lipids and cardiovascular disease risk biomarkers [39]. We used the serum samples to measure Eotaxin-1, and other SCI biomarkers and utilized the Inflammatory Age (iAge ® ) test (Edifice Health, Inc., San Mateo, CA, USA) as a study endpoint. ...
Article
Full-text available
Despite being largely preventable, cardiovascular disease (CVD) is still the leading cause of death globally. Recent studies suggest that the immune system, particularly a form of systemic chronic inflammation (SCI), is involved in the mechanisms leading to CVD; thus, targeting SCI may help prevent or delay the onset of CVD. In a recent placebo-controlled randomized clinical trial, an oat product providing 3 g of β-Glucan improved cholesterol low-density lipoprotein (LDL) levels and lowered cardiovascular risk in adults with borderline high cholesterol. Here, we conducted a secondary measurement of the serum samples to test whether the oat product has the potential to reduce SCI and improve other clinical outcomes related to healthy aging. We investigated the effects of the oat product on a novel metric for SCI called Inflammatory Age® (iAge®), derived from the Stanford 1000 Immunomes Project. The iAge® predicts multimorbidity, frailty, immune decline, premature cardiovascular aging, and all-cause mortality on a personalized level. A beneficial effect of the oat product was observed in subjects with elevated levels of iAge® at baseline (>49.6 iAge® years) as early as two weeks post-treatment. The rice control group did not show any significant change in iAge®. Interestingly, the effects of the oat product on iAge® were largely driven by a decrease in the Eotaxin-1 protein, an aging-related chemokine, independent of a person’s gender, body mass index, or chronological age. Thus, we describe a novel anti-SCI role for oats that could have a major impact on functional, preventative, and personalized medicine.
... A previous study demonstrated that dietary supplementation with β-glucan can improve cognitive function in diet-induced obese mice via the microbiota-gut-brain axis (Shi et al. 2020). Moreover, a recent randomized controlled trial reported that dietary β-glucan intake can reduce headache, fatigue, and anxiety in adults (Wolever et al. 2021). However, it is still unknown whether β-glucan administration can exert a protective effect on T. gondii-induced anxiety. ...
... Interestingly, several studies including ours have demonstrated that dietary supplementation with β-glucan can alleviate cognitive impairment via the microbiota-gut-brain axis (Pan et al. 2021;Shi et al. 2020). Importantly, in a recent randomized controlled trial, dietary β-glucan intake was reported to reduce headache, fatigue, and anxiety in adults (Wolever et al. 2021). Here, intraperitoneal administration of β-glucan alleviated the anxiety-like behavior induced by T. gondii infection and reduced the cyst burden in the brains of infected mice. ...
Article
Full-text available
Chronic Toxoplasma gondii (T. gondii) infection has been revealed to be a risk factor for neuropsychiatric diseases, including anxiety. However, there is no intervention strategy. The present study aimed to investigate the protective effect of β-glucan on T. gondii Wh6 strain–induced anxiety-like behavior in mice. The anxiety mouse model was established by infection with 10 cysts of the T. gondii Wh6 strain. β-Glucan was intraperitoneally administered 2 weeks before infection. Open field and elevated plus maze tests were performed to assess anxiety-like behavior. In the open field test, Wh6-infected mice spent less time in the central zone and had fewer entries into the central zone. In the elevated plus maze test, the infection reduced the frequency and time of head entries in the open arms. These results showed that Wh6 causes anxiety-like behavior in mice. Interestingly, the administration of β-glucan significantly ameliorated anxiety-like behavioral performance. The present study shows that β-glucan can alleviate the anxiety-like behavior induced by chronic T. gondii infection in mice, which indicates that β-glucan may be a potential drug candidate for treating T. gondii–related mental disorders, including anxiety.
... Studies have shown that β-glucan in oats can improve immunity and have some anticancer effects [21]. In addition, as a prebiotic, it can be fermented by Bifidobacteria and Lactobacilli in the intestinal tract, promoting the proliferation of these bacteria and thus affecting the intestinal tract [22][23][24]. The intestinal flora is regulated. ...
Article
Full-text available
Camel milk is a nutrient-rich diet and fermentation affects its nutritional value and probiotic function. In this study, sour camel milk and oat jujube sour camel milk were prepared using fermentation bacteria agent TR1, and the metabolites of camel milk, sour camel milk and oat jujube sour camel milk were detected using a non-targeted metabolomics approach using liquid chromatography-mass spectrometry (LC-MS).The results showed that the partial least squares discriminant analysis (PLS-DA) with 100 % accuracy and good predictive power detected 343 components in positive ion mode and 220 components in negative ion mode. The differential metabolites were mainly organic acids, amino acids, esters, vitamins and other substances contained in camel milk.It showed that there were significant differences in the metabolites of camel milk, sour camel milk and oat jujube sour camel milk. Based on the pathway enrichment analysis of the three dairy products in the KEGG database, 12 metabolic pathways mainly involved in the positive ion mode and 20 metabolic pathways mainly involved in the negative ion mode were identified. The main biochemical metabolic pathways and signal transduction pathways of the differential metabolites of the three dairy products were obtained. This study provides theoretical support for improving the nutritional quality and probiotic function of camel milk and fermented camel milk products and provides a basis for the development of relevant processing technologies and products for camel milk and fermented camel milk.
... Moreover, the European Food Safety Authority (ESFA) has approved health claims for cereal β-glucans related to a reduction in postprandial glycemic responses and serum cholesterol [3,4]. Additional studies suggest that ingestion of cereal β-glucan has other benefits such as glycemic control in type 1 diabetes [5], attenuating cognitive impairment [6], improving chronic kidney diseases [7], and reducing fatigue [8]. ...
Article
Full-text available
β-glucans found in cereal grains have been previously demonstrated to improve blood glucose control; however, current understanding points to their high viscosity as the primary mechanism of action. In this work, we present a novel, highly soluble, low-viscosity β-glucan fiber (HS-BG fiber) and a preclinical dataset that demonstrates its impact on two mechanisms related to the prevention of hyperglycemia. Our results show that HS-BG inhibits the activity of two key proteins involved in glucose metabolism, the α-glucosidase enzyme and the SGLT1 transporter, thereby having the potential to slow starch digestion and subsequent glucose uptake. Furthermore, we demonstrate in a multi-donor fecal fermentation model that HS-BG is metabolized by several different members of the gut microbiome, producing high amounts of short-chain fatty acids (SCFAs), known agonists of GPR43 receptors in the gut related to GLP-1 secretion. The production of SCFAs was verified in the translational gut model, SHIME®. Moreover, HS-BG fiber fermentation produces compounds that restored permeability in disrupted epithelial cells, decreased inflammatory chemokines (CXCL10, MCP-1, and IL-8), and increased anti-inflammatory marker (IL-10), which could improve insulin resistance. Together, these data suggest that the novel HS-BG fiber is a promising new functional ingredient that can be used to modulate postprandial glycemic responses while the high solubility and low viscosity enable easy formulation in both beverage and solid food matrices.
... The effects of probiotics on anxiety generally entail the hypothalamo-pituitary-adrenocortical (HPA) axis, intestinal permeability, and inflammation (Tremblay et al. 2021). In addition to probiotics, manipulations affecting anxiety comprise the use of germ-free mice, antibiotics, and prebiotics (Foster 2021), the latter defined as substances that stimulate the gut microbiota ) such as galacto-oligosaccharides (GOS), which decreased wake-up salivary cortisol levels in healthy subjects (Schmidt et al. 2015), and beta-glucan, whose reduction of self-reported anxiety symptoms in healthy subjects reached borderline significance (Wolever et al. 2021). ...
Article
Gut microbiota have been shown to be useful in treating gastrointestinal diseases, cancer, obesity, infections, and, more recently, neuropsychiatric conditions such as degenerative diseases and depression. There has also been recent expansion in testing probiotics and prebiotics on anxiety-like behaviors in animals. Current results indicate that probiotic substances of the Lactobacillus and Bifidobacterium type are effective in reducing anxiety-like behaviors in mice or rats evaluated in the elevated plus-maze, the open-field, the light-dark box, and conditioned defensive burying. Probiotics are also effective in reducing serum or plasma corticosterone levels after acute stress. It is hypothesized that probiotics cause anxiolytic-like effects via vagal influences on caudal solitary nucleus, periaqueductal gray, central nucleus of the amygdala, and bed nucleus of the stria terminalis. Further experimentation is needed to trace the neurochemical anatomy underlying anxiolytic-like behaviors of gut microbiata exerting effects via vagal or nonvagal pathways.
... Structural studies report that combinations of β-1,3 and β-1,6 bonds form long linear β-glucans, and these structures can be detected by specific intestinal receptors, such as dectin-1, which then stimulate the immunological system [1]. Cereal β-glucans that have been derived from barley and oats have also been widely researched in both animal and human studies [2][3][4][5]. They are water soluble, viscous polysaccharides with a linear structure in which glucose is bound through β-1,4 and β-1,3 linkages. ...
Article
Full-text available
Many articles and manuscripts focusing on the structure, function, mechanism of action, and effects of β-glucan have been published recently [...]
Article
Dairy products are a core part of many cultures' daily diets owing to their nutritional properties. The health benefits associated with dairy foods extend far beyond providing dietary needs, as dairy food consumption has been linked to a reduced risk of heart disease, stroke, hypertension, type 2 diabetes, metabolic syndrome, and colorectal cancer. However, lactose intolerance, milk protein allergies, cholesterol levels, and the emerging demand for vegetarianism have recently driven the need for non-dairy products. As a result, grain-based products are gaining popularity among consumers as a healthier alternative to dairy products. Thus, a new nutrition concept involves the development of new products that combine cereals and bioactive compounds to enhance the nutritional and distinctive characteristics of the product. Thus, these products' concomitant nutritional and bioactive composition confers a suitable profile for the transport and administration of probiotics by food. In addition, the presence of prebiotics in their natural configuration makes them useful to ensure the viability of probiotics and the bioavailability of bioactive compounds after their exposure to digestive conditions. However, incorporating probiotics and bioactive compounds such as fatty acids in a complex matrix is a technological challenge, mainly due to the low solubility of the cereals used in production. In this context, this review details the use of cereals as alternatives to dairy products in functional food formulation. It addresses the technological challenges of incorporating probiotics and bioactive compounds into these different matrices and the opportunities for their application in the food industry.
Article
Full-text available
Objectives The efficacy of oat beta-glucan (OBG), a viscous soluble fibre, on postprandial glycemic outcomes may depend on the nature of the control and the dose and molecular weight (MW) utilized. We undertook a systematic review and meta-analysis of acute clinical trials to determine whether these features mediate the glycemic and insulinaemic responses to OBG. Methods MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials were searched through October 27, 2020. We included acute, single-meal feeding, controlled trials investigating the effect of OBG (concentrate or oat bran) added to a carbohydrate-containing meal compared to a comparable meal (matched control) or a different carbohydrate-containing meal (unmatched control). Two reviewers extracted the data and assessed the risk of bias and certainty of evidence (GRADE). The primary outcome was incremental area under the curve (iAUC) for blood glucose. Data were pooled using the generic-inverse variance method with random effects model and expressed as ratio of means with [95% Cis]. Results One hundred and three trial comparisons (N = 538) were included. OBG reduced glucose iAUC and iPeak by 23% (0.77 [0.74, 0.81]) and 28% (0.72 [0.64, 0.76]) and insulin by 22% (0.78 [0.72, 0.85]) and 24% (0.76 [0.65, 0.88]), respectively. Dose, molecular-weight and comparator were significant effect modifiers of glucose iAUC and iPeak. Significant linear dose-response relationships were observed for all outcomes. OBG molecular-weight > 300 kg/mol significantly reduced glucose iAUC and iPeak, whereas, molecular-weight < 300 kg/mol did not. Reductions in glucose iAUC (27 vs 20%, p = 0.03) and iPeak (39 vs 25%, p < 0.01) were significantly larger with different vs comparable control-meals. Outcomes were similar in participants with and without diabetes. All outcomes had high certainty-of-evidence. Conclusions Current evidence indicates that the addition of OBG to carbohydrate-containing meals reduces the postprandial glycemic and insulinaemic responses. However, the magnitude of glucose reduction depends on OBG dose, molecular-weight and the comparator. Funding Sources INQUIS Clinical Research Ltd. (formerly GI Labs), and PepsiCo Global R&D
Article
Full-text available
Background High-molecular-weight (MW) oat β-glucan (OBG), consumed at 3–4 g/d, in solid foods reduces LDL cholesterol by a median of ∼6.5%. Objectives We evaluated the effect of a beverage providing 3 g/d high-MW OBG on reduction of LDL cholesterol (primary endpoint) when compared with placebo. Methods We performed a parallel-design, randomized clinical trial at a contract research organization; participants, caregivers, and outcome assessors were blinded to treatment allocation. Participants with LDL cholesterol between 3.0 and 5.0 mmol/L, inclusive [n = 538 screened, n = 260 ineligible, n = 23 lost, n = 48 withdrawn (product safety); n = 207 randomly assigned, n = 7 dropped out, n = 9 withdrawn (protocol violation); n = 191 analyzed; n = 72 (37.7%) male, mean ± SD age: 43.3 ± 14.3 y, BMI: 29.7 ± 5.2 kg/m2], were randomly assigned to consume, 3 times daily for 4 wk, 1 g OBG (n = 104, n = 96 analyzed) or rice powder (Control, n = 103, n = 95 analyzed) mixed into 250 mL water. Treatment effects were assessed as change from baseline and differences analyzed using a 2-sided t test via ANOVA with baseline characteristics as covariates. Results After 4 wk, change from baseline least-squares-mean LDL cholesterol on OBG (−0.195 mmol/L) was less than on Control (0.012 mmol/L) by mean: 0.207 mmol/L (95% CI: 0.318, 0.096 mmol/L; P = 0.0003); the following secondary endpoints were also reduced as follows: total cholesterol (TC) (0.226 mmol/L; 95% CI: 0.361, 0.091 mmol/L; P = 0.001), TC:HDL cholesterol ratio (0.147; 95% CI: 0.284, 0.010; P = 0.036), non-HDL cholesterol (0.194 mmol/L; 95% CI: 0.314, 0.073 mmol/L; P = 0.002), and Framingham cardiovascular disease (CVD) risk (0.474; 95% CI: 0.900, 0.049, P = 0.029). Changes in HDL cholesterol, triglycerides, glucose, and insulin did not differ between treatment groups (P > 0.05). Lipid treatment effects were not significantly modified by age, sex, BMI, or hypertension treatment. There were no major adverse events, but both treatments transiently increased gastrointestinal symptoms. Conclusions Consuming a beverage containing 1 g high-MW OBG 3 times daily for 4 wk significantly reduced LDL cholesterol by ∼6% and CVD risk by ∼8% in healthy adults with LDL cholesterol between 3 and 5 mmol/L. This trial was registered at clinicaltrials.gov as NCT03911427.
Article
Full-text available
To determine the effect of oat β‑glucan (OBG) on acute glucose and insulin responses and identify significant effect modifiers we searched the MEDLINE, EMBASE, and Cochrane databases through October 27, 2020 for acute, crossover, controlled feeding trials investigating the effect of adding OBG (concentrate or oat-bran) to carbohydrate-containing test-meals compared to comparable or different carbohydrate-matched control-meals in humans regardless of health status. The primary outcome was glucose incremental area-under-the-curve (iAUC). Secondary outcomes were insulin iAUC, and glucose and insulin incremental peak-rise (iPeak). Two reviewers extracted the data and assessed risk-of-bias and certainty-of-evidence (GRADE). Data were pooled using generic inverse-variance with random-effects model and expressed as ratio-of-means with [95% CIs]. We included 103 trial comparisons ( N = 538). OBG reduced glucose iAUC and iPeak by 23% (0.77 [0.74, 0.81]) and 28% (0.72 [0.64, 0.76]) and insulin by 22% (0.78 [0.72, 0.85]) and 24% (0.76 [0.65, 0.88]), respectively. Dose, molecular-weight, and comparator were significant effect modifiers of glucose iAUC and iPeak. Significant linear dose-response relationships were observed for all outcomes. OBG molecular-weight >300 kg/mol significantly reduced glucose iAUC and iPeak, whereas molecular-weight <300 kg/mol did not. Reductions in glucose iAUC (27 vs 20%, p = 0.03) and iPeak (39 vs 25%, p < 0.01) were significantly larger with different vs comparable control-meals. Outcomes were similar in participants with and without diabetes. All outcomes had high certainty-of-evidence. In conclusion, current evidence indicates that adding OBG to carbohydrate-containing meals reduces glycaemic and insulinaemic responses. However, the magnitude of glucose reduction depends on OBG dose, molecular-weight, and the comparator.
Article
Full-text available
Abstract Background In vitro and animal experiments reported a microbiota-regulating ability of oatmeal, however, related in vivo evidences remained limited. Thus, we conducted this study aiming to investigate the oatmeal-induced alteration of gut microbiota and its potential relationship with the improvements of lipid profiles. Methods and study design Data of anthropometric measurements and biochemical parameters were extracted from a randomized, controlled clinical trial, in which 62 hypercholesterolemic men and women (18–65 years old) were provided with either treatment of 80 g/day oatmeal or 80 g/day refined white rice for 45 days. Fasting blood samples and fecal samples were collected both at baseline and endpoint of the study for lipid profiling and microbiota 16S rRNA amplicon sequencing, respectively. Results Totally 28 participants (56 fecal samples) qualified with the new criteria and were thus included in this secondary analysis. The results of microbiota analysis showed that no significant difference was observed in the alteration of its overall α or β diversity between two groups throughout the study. Nor did any notable between-group difference was found in the relative abundance changes of microorganism at different taxonomies. However, results from linear discriminant analysis effect size in the oatmeal group indicated a significant positive response of Firmicutes phylum following oatmeal consumption. Further Procrustes analysis suggested a concordance trend between microorganism alteration and alleviation of hypercholesterolemia phenotypes throughout the study (P = 0.05). The results of within-group comparison from Spearman’s correlation in the oatmeal group demonstrated a significant association between the enrichment of Blautia genus and the reduction of serum total cholesterol (P
Article
Full-text available
Published meta-analyses indicate significant but inconsistent incident type-2 diabetes(T2D)-dietary glycemic index (GI) and glycemic load (GL) risk ratios or risk relations (RR). It is nowover a decade ago that a published meta-analysis used a predefined standard to identify validstudies. Considering valid studies only, and using random effects dose-response meta-analysis(DRM) while withdrawing spurious results (p < 0.05), we ascertained whether these relationswould support nutrition guidance, specifically for an RR > 1.20 with a lower 95% confidence limit>1.10 across typical intakes (approximately 10th to 90th percentiles of population intakes). Thecombined T2D-GI RR was 1.27 (1.15-1.40) (p < 0.001, n = 10 studies) per 10 units GI, while that forthe T2D-GL RR was 1.26 (1.15-1.37) (p < 0.001, n = 15) per 80 g/d GL in a 2000 kcal (8400 kJ) diet.The corresponding global DRM using restricted cubic splines were 1.87 (1.56-2.25) (p < 0.001, n =10) and 1.89 (1.66-2.16) (p < 0.001, n = 15) from 47.6 to 76.1 units GI and 73 to 257 g/d GL in a 2000kcal diet, respectively. In conclusion, among adults initially in good health, diets higher in GI or GLwere robustly associated with incident T2D. Together with mechanistic and other data, thissupports that consideration should be given to these dietary risk factors in nutrition advice.Concerning the public health relevance at the global level, our evidence indicates that GI and GLare substantial food markers predicting the development of T2D worldwide, for persons ofEuropean ancestry and of East Asian ancestry.
Article
Full-text available
Aim The aim of this study was to investigate if working in a cold environment and feeling cold at work are associated with chronic pain (ie, lasting ≥3 months). Methods We used data from the sixth survey (2007–2008) of the Tromsø Study. Analyses included 6533 men and women aged 30–67 years who were not retired, not receiving full-time disability benefits and had no missing values. Associations between working in a cold environment, feeling cold at work and self-reported chronic pain were examined with logistic regression adjusted for age, sex, education, body mass index, insomnia, physical activity at work, leisure time physical activity and smoking. Results 779 participants reported working in a cold environment ≥25% of the time. This exposure was positively associated with pain at ≥3 sites (OR 1.57; 95% CI 1.23 to 2.01) and with neck, shoulder and leg pain, but not with pain at 1–2 sites. Feeling cold sometimes or often at work was associated with pain at ≥3 sites (OR 1.58; 95% CI 1.22 to 2.07 and OR 3.90; 95% CI 2.04 to 7.45, respectively). Feeling cold often at work was significantly and positively associated with pain at all sites except the hand, foot, stomach and head. Conclusion Working in a cold environment was significantly associated with chronic pain. The observed association was strongest for pain at musculoskeletal sites and for those who often felt cold at work.
Article
Full-text available
Objective To assess the association between hot flashes (HFs) severity and oxidative stress (OS) in Mexican postmenopausal women. Methods A cross-sectional study was carried out with perimenopausal women aged 40–59 years community-dwelling from Mexico City, Mexico. They participated in Menopause and Oxidative Stress Project. The baseline sample consisted of 476 women recruited to participate; 161 women were excluded due to different reasons. Hence, 315 women were selected to establish two groups, a) 145 premenopausal women (yet with menstrual bleeding), and b) 170 postmenopausal women (without menses). All women were free of cardiovascular, kidney, hepatic or cancer disease, and without antioxidant supplement intake for at least six months prior to the beginning of the study; none had previously received hormone therapy. As OS markers, we measured plasma malondialdehyde using the TBARS assay, erythrocyte superoxide dismutase (SOD) and glutathione peroxidase (GPx), uric acid, and total antioxidant status; also, we calculated SOD/GPx ratio, antioxidant gap and an oxidative stress score ranging from 0 to 7. The HFs were evaluated using the Menopause Rating Scale. The women completed Spanish version of the Athens Insomnia Scale, Zung Self-Rating Anxiety Scale and Zung Self-Rating Depression Scale and a questionnaire of pro-oxidant factors. Results Stress score increased with HFs severity (mild 2.7±0.17, moderate 2.9±0.20 and severe 3.7±0.20, p = 0.001) in postmenopausal women. We observed a positive correlation between HFs severity and stress score, r = 0.247 (p = 0.001) in postmenopausal women; other test scores were not correlated. Severe HFs were a risk factor for OS (OR = 5.12, 95%CI: 1.99–13.17, p<0.05) in an adjusted multivariate analysis by different postmenopausal symptoms and pro-oxidant factors; we did not see any association in premenopausal women. Conclusion Our findings suggest an association between HFs severity and OS in Mexican postmenopausal women.
Article
In a striking display of trans-kingdom symbiosis, gut bacteria cooperate with their animal hosts to regulate the development and function of the immune, metabolic and nervous systems through dynamic bidirectional communication along the ‘gut–brain axis’. These processes may affect human health, as certain animal behaviours appear to correlate with the composition of gut bacteria, and disruptions in microbial communities have been implicated in several neurological disorders. Most insights about host–microbiota interactions come from animal models, which represent crucial tools for studying the various pathways linking the gut and the brain. However, there are complexities and manifest limitations inherent in translating complex human disease to reductionist animal models. In this Review, we discuss emerging and exciting evidence of intricate and crucial connections between the gut microbiota and the brain involving multiple biological systems, and possible contributions by the gut microbiota to neurological disorders. Continued advances from this frontier of biomedicine may lead to tangible impacts on human health.
Article
Dietary fiber is a crucial component of a healthy diet, with benefits that can be attributed to processes in the gut microbiota and the resulting by-products. Observational studies support associations between dietary fiber intake and depression and inflammation, but the potential mechanisms are poorly understood. This review examines evidence of the effects of dietary fiber on depression and inflammation and considers plausible mechanisms linking dietary fiber and depression, including microbiota-driven modification of gene expression and increased production of neurotransmitters. Additionally, inflammation may mediate the relationship between dietary fiber intake and depression. A high-fiber diet potentially lowers inflammation by modifying both the pH and the permeability of the gut. The resultant reduction in inflammatory compounds may alter neurotransmitter concentrations to reduce symptoms of depression. Further research into the link between dietary fiber intake and inflammation and depression is essential, as findings could potentially provide guidance for improvement in or prevention of inflammatory and depressive disorders.
Article
Oats are uniquely nutritious, owing to their composition of bioactive compounds, lipids, and β-glucan. Scientific research has established that oats can improve diet quality, reduce cholesterol, regulate satiety, and protect against carcinogenesis in the colon; however, determining the effects of oats on gastrointestinal health and the gut microbiome is a newer, evolving area of research. To better understand the effects of oats on gastrointestinal health in humans, a literature review with predefined search criteria was conducted using the PubMed database and keywords for common gastrointestinal health outcomes. Moreover, to examine the gastrointestinal effects of oats across the scientific spectrum, a similar search strategy was executed to identify animal studies. In vitro studies were identified from the reference lists of human and animal studies. A total of 8 human studies, 19 animal studies, and 5 in vitro studies met the inclusion criteria for this review. The evidence in humans shows beneficial effects of oats on gastrointestinal health, with supportive evidence provided by in vitro and animal studies. The effective dose of oats varies by type, although an amount providing 2.5 to 2.9 g of β-glucan per day was shown to decrease fecal pH and alter fecal bacteria. For oat bran, 40 to 100 g/d was shown to increase fecal bacterial mass and short-chain fatty acids in humans. Differences in study design, methodology, and type of oats tested make valid comparisons difficult. The identification of best practices for the design of oat studies should be a priority in future research, as the findings will be useful for determining how oats influence specific indices of gastrointestinal health, including the composition of the human gut microbiome.