liu-et-al-2021-consumption-of-dietary-fiber-from-different-sources-during-pregnancy-alters-sow-gut-microbiota-and
liu-et-al-2021-consumption-of-dietary-fiber-from-different-sources-during-pregnancy-alters-sow-gut-microbiota-and
Host-Microbe Biology
College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
a
b Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
Boshuai Liu and Xiaoyan Zhu contributed equally to this article. Author order was determined alphabetically.
S ows and piglets are excellent animal models and have been widely used in bio-
medical research. Compared to rodents, sows and piglets are considered a superior
model for studying the relationships in gut function of human mothers and infants (1).
Pigs have many characteristics similar to humans, including digestive physiology,
microbiota, and diet. These animals are suitable for a multitude of disease models,
including diarrhea, gastrointestinal inflammatory disorders, necrotizing enterocolitis of
neonates, and obesity, etc. (2). In large-scale pig production, sows and piglets are cru-
cial to determining production levels and the economic benefits of pig farms, and the
gestational, lactation, and newborn periods are core stages for feed management of
sows and piglets in large-scale pig production (3). During pregnancy, sows undergo
dramatic changes in physiological metabolism and immunity to ensure the implanta-
tion and development of embryos and pregnancy completion (4). In the mid and late
periods of pregnancy, the levels of tumor necrosis factor alpha (TNF-a), interleukin-6
(IL-6), reactive oxygen species (ROS), and other proinflammatory factors increase signif-
icantly in the blood of sows (5) and are closely related to numerous diseases, including
constipation, abortion, and intrauterine growth retardation (IUGR) (6, 7). Further,
expression of the tight-junction protein, zonulin, is increased in sow guts, while bacte-
rial lipopolysaccharide (LPS) entering the circulation through the gut barrier increased,
and increased concentrations of bacterial endotoxins in the circulation can lead to
metabolic endotoxemia, which is a potential mediator of inflammation (8–10). To initi-
ate and maintain lactation, sows undergo complex metabolism and immune system
changes that directly affect the development and growth of piglets (11). Therefore,
reduction of inflammatory responses and ensuring normal metabolic and immune
changes in sows during mid and late pregnancy and lactation is crucial for the perform-
ance of sows and their offspring (4). Gut microbiota has key roles in nutrient metabolism,
immune development, protection against pathogens, and the pathogenesis of many
chronic diseases in the host (12–14). Sow gut microbiota change dramatically during
pregnancy and may be involved in metabolic processes in pregnant animals (15).
Compared to the early stage of pregnancy, Proteobacteria and Actinobacteria are signifi-
cantly increased in the sow gut during the late stage of pregnancy and have clear char-
TABLE 1 Effects of different fiber sources on fat thickness and feed intake of sows
Mean ± SDa
Description CK AM BP SH
Day 60 in gestation (mm) 19.82 6 1.00 19.83 6 1.35 19.83 6 1.16 19.68 6 0.84
Day 90 in gestation (mm) 19.54 6 0.78 19.77 6 1.22 19.83 6 1.08 19.73 6 0.61
Before parturition (mm) 20.84 6 2.37 20.43 6 1.43 20.72 6 1.35 19.77 6 0.80
After weaning (mm) 19.38 6 1.54 20.53 6 0.94 19.67 6 2.11 19.15 6 2.23
Gain during gestation (mm) 1.12 6 1.76 0.65 6 0.74 0.95 6 2.83 0.22 6 1.01
Loss during lactation (mm) 1.43 6 3.78 –0.09 6 0.16 1.20 6 1.87 0.62 6 2.44
Pregnancy feed intake (kg) 2.44 6 0.27 2.37 6 0.26 2.35 6 0.39 2.46 6 0.38
Lactation feed intake (kg) 7.75 6 0.66B 8.76 6 0.44A 7.80 6 0.64B 7.66 6 0.59B
aData for the control (CK), alfalfa meal (AM), beet pulp (BP), and soybean skin (SH) groups are presented. The
data were evaluated by one-way ANOVA, and significant differences between means were assessed by using
Duncan’s test. Differences in the superscript letters for peer data indicate that a difference is significant (P ,
0.05). The lack of a superscript letter means that all differences were nonsignificant (P . 0.05).
The effects of dietary fiber vary greatly due to their different sources and the variety
and complexity of their chemical structures (26). Darroch et al. (27) added 20% soy-
bean hulls and 0.3% psyllium to the diets of pregnant sows. Their results showed that
soybean hull was more conducive to physical health maintenance in pregnant sows
but had little effect on litter size. Cheng et al. (20) added combined soluble fiber from
pregelatinized waxy corn starch and guar gum to the sows’ pregnancy diet, which sig-
nificantly improved the developmental growth performance and gut function of 14-
day-old suckling piglets. Zhuo et al. (28) found that insoluble fiber oat bran mixed with
corn or soybean meal produced more SCFA by gut fermentation, which improved pig
behaviors and reproductive performance. Although the direct effects of dietary fiber
on gut microbiota composition have been studied extensively, systematic evaluation
of different fiber sources (e.g., insoluble and soluble fiber sources) on gut health and
inflammatory responses of sows and their offspring during mid and late gestation pe-
riod has rarely been conducted. In this study, we evaluated the effects of adding differ-
ent fiber sources, including soybean husk (SH), alfalfa meal (AM), and beet pulp (BP), to
RESULTS
Effects of supplementation in mid to late pregnancy with dietary fiber from
different sources on the performance of sows and piglets. Sows (n = 48; Large
Landrace) at 60 days of gestation were randomly allocated to groups as follows: control
animals (CK) and animals with dietary supplementation using alfalfa meal (AM), beet
pulp (BP), and soybean skin (SH). Assessment of sow backfat thickness during the
reproductive cycle, including at gestation day 60 (G60d), G90d, lactation day 0 (L0d),
and L21d, and sow feed intake by one-way analysis of variance (ANOVA) (Table 1)
showed that there was no significant difference in average daily feed intake (P =
0.7620) and backfat (P = 0.6290 and P = 0.4240, respectively) among the treatment
groups in mid to late pregnancy; however, in the lactation period, the average daily
feed intake (P = 0.0320) of the AM group was significantly higher than that of CK, BP,
and SH groups; in addition, the backfat loss of lactating sows showed a downward
trend (P = 0.3750). Evaluation of sow reproductive performance demonstrated no sig-
nificant differences in total litter size (P = 0.1380), live litter size (P = 0.1510), newborn
body weight (P = 0.4430), or newborn litter weight (P = 0.1170) among the treatment
groups; however, IUGR was significantly lower in the AM group than in the CK, BP, and
SH groups (P = 0.0370) (Table 2). Further, we conducted a systematic study of piglet
growth performance during lactation (L7d, L14d, and L21d) and found that individual
body weight (P = 0.0700, P = 0.1580, and P = 0.0980, respectively) and litter weight
Description CK AM BP SH
Total born piglets 13.11 6 2.85 12.13 6 3.31 10.44 6 4.28 13.11 6 2.62
Live-born piglets 12.67 6 2.83 12.00 6 3.42 10.11 6 4.28 12.56 6 2.46
IUGR (%) 9.48 6 0.07A 2.08 6 0.04B 11.21 6 0.08A 8.94 6 0.06A
Body wt of newborn piglets (kg) 1.35 6 0.17 1.37 6 0.12 1.40 6 0.17 1.42 6 0.17
Litter wt at birth (kg) 16.81 6 2.42 16.49 6 4.86 14.04 6 5.73 17.72 6 3.79
aData for the control (CK), alfalfa meal (AM), beet pulp (BP), and soybean skin (SH) groups are presented. The
data were evaluated by one-way ANOVA, and significant differences between means were assessed by using
Duncan’s test. Differences in superscript letters for peer data indicate that a difference is significant (P , 0.05).
The lack of a superscript letter means that all differences were nonsignificant (P . 0.05).
(P = 0.0014, P = 0.1580, and P = 0.0980, respectively) of piglets in the AM, BP, and SH
groups showed an increasing trend compared to the CK (Table 3). These data indicate
that AM intake during mid to late pregnancy can increase feed intake and reduce IUGR
in lactating sows.
Effects of AM diet on gut permeability and inflammatory responses in sows
and piglets. Given our findings that AM supplementation could reduce IUGR, we next
conducted a systematic evaluation of three biomarkers related to gut permeability
(ROS, endotoxin, and zonulin) by one-way ANOVA at G100d, L4d, and L18d in sows
with dietary supplementation with AM compared to controls. Compared to CK sows,
the ROS (P = 0.0004, P , 0.0001, and P = 0.0010, respectively) and endotoxin (P =
0.0100, P = 0.0018, and P = 0.0007, respectively) levels were significantly lower at
G100d, L4d, and L18d in serum samples from sows fed an AM diet (Fig. 1A and C).
Further, serum levels of zonulin at L18d were significantly lower than those in the CK
(P = 0.0014) (Fig. 1B). Together, these data suggest that AM intake in mid to late preg-
nancy reduces sow gut permeability. Next, we examined four biomarkers associated
with gut inflammation in sows, IL-6, lipocalin-2, TNF-a, and IL-10. In terms of systemic
inflammatory responses, we found that serum levels of IL-6 (P = 0.0014, P = 0.0010,
and P , 0.0001, respectively), lipocalin-2 (P = 0.0122, P = 0.0039, and P = 0.0044,
Description CK AM BP SH
Litter wt (kg)
Day 0 19.48 6 2.24 19.42 6 2.02 19.44 6 2.89 19.53 6 1.23
Day 7 39.82 6 1.74 39.87 6 4.33 43.53 6 4.61 42.76 6 4.16
Day 14 54.90 6 3.77 54.91 6 2.38 57.59 6 5.25 57.51 6 6.39
Day 21 76.16 6 5.80 82.56 6 1.15 79.64 6 6.66 75.93 6 7.60
Avg daily gain 2.83 6 0.29 3.16 6 0.09 3.01 6 0.20 2.82 6 0.34
Body wt (kg)
Day 0 1.38 6 0.20 1.38 6 0.18 1.38 6 0.26 1.39 6 0.11
Day 7 3.62 6 0.16 3.62 6 0.39 3.96 6 0.42 3.89 6 0.38
Day 14 4.99 6 0.34 4.99 6 0.22 5.24 6 0.48 5.00 6 0.66
Day 21 6.92 6 0.53 7.51 6 0.10 7.24 6 0.61 6.90 6 0.69
Avg daily gain 0.26 6 0.03 0.29 6 0.01 0.27 6 0.01 0.26 6 0.03
aData for the control (CK), alfalfa meal (AM), beet pulp (BP), and soybean skin (SH) groups are presented. Eleven
animals were included in each group. The data were evaluated by one-way ANOVA, and significant differences
between means were assessed by using Duncan’s test. Differences in superscript letters for the peer data
indicate that a difference is significant (P , 0.05). The lack of a superscript letter means that all differences were
nonsignificant (P . 0.05).
FIG 1 Effect of alfalfa meal diet on serum ROS (A), serum zonulin (B), serum endotoxin (C), serum
lipocalin-2 (D), serum IL-6 (E), serum TNF-a (F), serum IL-10 (G), fecal endotoxin (H), fecal IL-6 (I), fecal
TNF-a (J), and fecal IL-10 (K) levels of sows. CK, control group; AM, alfalfa meal group. The data were
evaluated by one-way ANOVA, and significant differences between means were assessed by using
Duncan’s test. *, 0.01 , P # 0.05; **, 0.001 , P # 0.01; ***, P # 0.001; ns . 0.05.
FIG 2 Effect of alfalfa meal diet on serum ROS (A), serum endotoxin (B), serum IL-6 (C), serum TNF-a (D) and
serum IL-10 (E) levels of piglets. CK, control group; AM, alfalfa meal group. The data were evaluated by one-
way ANOVA, and significant differences between means were assessed by using Duncan’s test. *, 0.01 , P #
0.05; **, 0.001 , P # 0.01; ***, P # 0.001; ns, P . 0.05.
demonstrated that levels of IL-6 (P = 0.0010 and P = 0.0017, respectively) and TNF-a
(P = 0.0052 and P = 0.0084, respectively) were significantly reduced at L4d and L18d in
fecal samples from sows fed with an AM, while those of IL-10 (P = 0.0130 and P =
FIG 3 AM diet regulates the changes of gut microbiota composition in sows. (A) Principal-component analysis of OTU level by
Bray-Curtis and unweighted-UniFrac distance; (B) LEfSe analysis determined by one-against-all (less strict). CK, control group;
AM, alfalfa meal group.
FIG 4 Effect of alfalfa meal diet on SCFAs fermentation of sows. (A) Fecal amino acids (AA); (B) fecal propionic
acid (PA); (C) fecal butyric acid (BA); (D) fecal total SCFAs. The data were evaluated by one-way ANOVA, and
the significant differences between means were assessed by using Duncan’s test. *, 0.01 , P # 0.05; **,
0.001 , P # 0.01; ***, P # 0.001; ns, . 0.05.
size (LEfSe) analysis by “one-against-all” (less strict) to evaluate differences between the
two groups showed that, at G100d, there was a significant increase in the relative abun-
TABLE 4 Correlation between IUGR, lactation feed intake, and inflammatory factors of sows
Correlationa
Description Serum lipocalin-2 Serum IL-6 Serum TNF-a Serum IL-10 Fecal IL-6 Fecal TNF-a Fecal IL-10 Serum ROS
IUGR (L4d) 0.864* 0.914* 0.926** –0.923** 0.773 0.363 –0.616 0.840
Lactation feed intake (L18d) –0.649* –0.638* –0.695* 0.685* –0.745** –0.398 0.201 –0.806**
aThedata were evaluated by Spearman correlation analysis of the Euclidean distance. *, Mean significant correlation (P , 0.05); **, mean extremely significant correlation
(P , 0.01).
significantly negatively correlated with sow fecal TNF-a. Further, piglet serum TNF-a
levels were positively correlated with serum IL-6, TNF-a, and ROS, as well as fecal IL-6
in sows, and significantly negatively correlated with piglet weaning body weight.
Serum IL-6 in piglets was positively correlated with serum lipocalin-2, IL-6, TNF-a, and
ROS in sows, while there was no significant correlation between serum ROS in piglets
and sow inflammatory factors or piglet weaning body weight.
The composition of gut microbiota of sows regulated by an AM supplemented
diet is related to their gut health. As shown in Fig. 5A, analysis of the correlation
between microbiota differing according to LEfSe and metabolic indices in the gut tract
of sows at each stage showed that the Prevotellaceae_NK3B31_group was significantly
positively correlated with serum IL-10, while the norank_f_2534-18b5_gut_group was
significantly positively correlated with serum IL-10 and fecal IL-10 and negatively corre-
lated with serum lipocalin-2, IL-6, and TNF-a. Terrisporobacter was positively correlated
with serum lipocalin-2, TNF-a, and endotoxin and negatively correlated with serum IL-
10. The Lachnospiraceae_NK4A136_group was positively correlated with serum IL-10,
and Clostridium_sensu_stricto_1 was significantly negatively correlated with serum zon-
ulin and ROS levels. The Ruminococcaceae_V9D2013_group was positively correlated
with serum zonulin and ROS and negatively correlated with serum IL-10, and the
Eubacterium_fissicatena_group was positively correlated with serum zonulin, ROS, TNF-a,
and endotoxin, and fecal IL-6. The Norank_f_2534-18b5_gut_group was significantly posi-
tively correlated with fecal IL-6 and negatively correlated with fecal IL-10. There were sig-
nificant positive correlations between unclassified_f_lachnospiraceae and serum zonulin
TABLE 5 Correlation between inflammatory factors of piglets and sows and weaning weight (L18d)
Correlation (sow group)a
Piglet group Serum lipocalin-2 Serum IL-6 Serum TNF-a Serum IL-10 Fecal IL-6 Fecal TNF-a Fecal IL-10 Serum ROS 21d wt
Serum IL-10 –0.547 –0.698 –0.589 0.836* –0.809 –0.885* 0.799 –0.630 0.324
Serum TNF-a 0.705 0.887* 0.877* –0.691 0.822* 0.750 –0.693 0.832* –0.866*
Serum IL-6 0.951** 0.910* 0.900* 20.627 0.69 0.712 20.735 0.825* 20.730
Serum ROS 0.537 0.719 0.661 20.467 0.635 0.517 20.605 0.746 20.662
aThedata were evaluated by Spearman correlation analysis of the Euclidean distance. *, Mean significant correlation (P , 0.05); **, mean extremely significant correlation
(P , 0.01).
DISCUSSION
The immune health status of reproductive sows directly affects overall pig produc-
tivity (3). During pregnancy, sow metabolism is enhanced, which manifests as an
increase in appetite, digestive capacity, weight gain, and storage of numerous
nutrients, to meet the requirements of fetal development. In the later stages of preg-
nancy, in addition to dietary energy, sows cease fat deposition and mobilize stored fat
and energy during pregnancy, which is transferred to the mammary gland for milk syn-
thesis. The metabolism and immune changes in sows during pregnancy and lactation
affect the development and growth of fetuses, and disruption of these adaptive
changes may lead to premature birth or even abortion (4). According to statistical anal-
yses, in commercial genetic lines, the prebirth loss of piglets is around 30 to 50% (29).
Therefore, it is crucial to reduce inflammatory responses in sows during mid and late
pregnancy and lactation and to ensure that the normal metabolic immune changes
occur in the sow reproductive cycle.
Some studies have shown that dietary fiber supplementation not only promotes
satiety in sows but also improves sow feed intake during lactation (28, 30, 31); how-
ever, the effects of dietary fiber supplementation from different sources are inconsis-
tent and may be closely related to the physicochemical properties and fermentability
of dietary fiber (20, 27, 28, 32, 33). Here, we found that the addition of different fiber
sources to the diet of sows in mid and late gestation affected the performance of both
sows and piglets, with AM in particular significantly reducing the IUGR rate, increasing
feed intake during lactation, and improving sow and piglet performance. AM is rich in
insoluble fiber but also contains a small amount of soluble fiber, which can be fer-
mented in the foregut segment, while the insoluble fiber can be slowly fermented in
the hindgut, and has beneficial effects throughout the intestine (34). Dietary fiber can
prevent and treat gut inflammation induced by a high-carbohydrate and low-fiber
western diet in mice by restoring the damaged gut mucous layer (35). In the case of
long-term or indirect dietary fiber deficiency, the gut microbiota resort to the use of
mucosal glycoprotein secreted by the host as a nutrition source, leading to the erosion
of the mucosal barrier. This results in increased pathogenic bacterial invasion of the
mucosa and aggravation of physiological inflammatory responses (36). Prefeeding
mice with inulin can reduce the gut inflammatory response and Smad7 expression af-
ter infection with Citrobacter rodentium and promote host protective immune
responses by affecting the NF-κB and Smad7 signal transduction pathways (37); how-
ever, longitudinal studies on the dynamic changes in inflammation in sows during mid
and late pregnancy and lactation are insufficient. In a recent study, sows that were in
mid and late gestation and lactating were found to have symptoms of metabolic syn-
drome, mainly characterized by low-level inflammation and metabolic disorder (38).
We assessed the gut permeability, gut and systemic inflammatory responses, and met-
abolic changes in sows fed AM. During the middle and late gestation and lactation
periods in sows fed with AM, we found that three biomarkers of gut permeability (ROS,
endotoxin, and connexin) and markers of inflammation (IL-6, lipocalin-2, and TNF-a)
were decreased, which indicated that AM in the sow’s diet reduced gut permeability
and decreased gut and systemic inflammation. Further, Spearman correlation analysis
showed that there was a significant positive correlation between IL-10 in piglets and
adjusting gut bacteria. Dietary fiber can not only regulate gut microbiota composition
but also adjust microbial metabolites, including SCFAs, etc. Further, it can improve gut
health and influence metabolism and animal behavior (41, 42). Zhao et al. reported
that high dietary fiber could enrich 15 SCFAs produced by bacteria in the gut, stimu-
late SCFA production, improve the gut environment, reduce gut pH, increase butyrate
concentration, competitively inhibit other “harmful bacterial,” and reduce the produc-
tion of harmful metabolites (such as indoles and hydrogen sulfide) and thus build a
healthier gut environment (43). Patients with irritable bowel syndrome have abnormal
gut microbiota due to insufficient intake of SCFAs by gut epithelial cells, and the distri-
bution of tight-junction proteins is directly affected, resulting in thinning of the gut
microbiota, increased gut permeability, and decreased protective effects (44, 45). It has
been suggested that dietary fiber-mediated changes in the gut microbiota and their
metabolites may have important roles in maintaining a gut microecological balance and
ensuring gut health. Our previous study (46) found that adding 5% AM to piglet diet
could inhibit harmful bacteria, such as Mycoplasma and Helicobacter in piglet guts, and
promote the proliferation of beneficial bacteria, such as Paenibacillus, Lactococcus,
Enterococcus, and Faecalibacterium. Compared to the nonpregnant period, the physio-
logical metabolic processes and immune systems of sows exhibit various changes in
pregnancy to meet their physiological needs during this period. The gut microbiota
changes significantly in different periods of pregnancy, which influences host metabolic
processes, and the changes are related to the metabolic characteristics and immune sys-
tem responses specific to the pregnancy period (47). Here, LEfSe analysis showed that
the addition of AM to sow diets in mid and late pregnancy significantly increased the
relative abundances of anti-inflammatory bacteria (Prevotellaceae_NK3B31_group,
norank_f_p_2534_18B5_gut_group, Lachnospiraceae_NK4A136_group, and g_Clostridium_
sensu_stricto_1) and decreased the relative abundance of proinflammatory bacteria
(Terrisporobacter, Desulfovibrio, Helicobacter, Eubacterium_fissicatena_group, and
Erysipelotrichaceae_UCG_004). Previous studies have found that colonization of
Clostridium_sensu_stricto_1 and other bacteria can promote the aggregation of CD41
regulatory T cells in the colon of sterile mice and improve the level of transforming
FIG 6 Systematic analysis of the effects of alfalfa meal diet on growth performance, inflammatory indexes, gut microbiota, and SCFAs of sows and piglets.
Terrisporobacter bacteria. Serum ROS levels were significantly positively correlated with
proinflammatory factors and endotoxins but negatively correlated with anti-inflamma-
tory factors, and there was a significant positive correlation between serum endo-
toxin and proinflammatory factors, as well as a significant positive correlation
during mid and late gestation influenced the performance of sows and piglets. In par-
ticular, the addition of AM significantly improved sow and piglet performance and
relieved gut and systemic inflammation. Furthermore, the supplementation with AM
significantly increased the relative abundance of anti-inflammatory bacteria and
decreased that of proinflammatory bacterial. We propose that the improvement in the
performance of sows and piglets can be ascribed to the beneficial effects of AM on gut
microbiota and the SCFA generation, resulting in decreased inflammatory responses
and enhanced physical health in sows and piglets. These findings provide a theoretical
basis and guide for the use of specific fiber sources in the diet of sows to improve gut
health and production performance of sows and piglets. Our data also give insights for
the study of the role of dietary fiber in the gastrointestinal function of human mothers
and infants.
(2 300 bp) on an Illumina MiSeq platform (Illumina, San Diego, CA), according to standard protocols,
by Majorbio Bio-Pharm Technology Co., Ltd. (Shanghai, China).
Bioinformatics analysis of sequencing data. Raw fastq files were demultiplexed, quality filtered
using Trimmomatic, and merged using FLASH, according to the following criteria: (i) reads were trun-
cated at any site receiving an average quality score of ,20 over a 50-bp sliding window; (ii) primers
were exactly matched, allowing 2-nucleotide mismatching, and reads containing ambiguous bases
removed; and (iii) sequences whose overlap was longer than 10 bp were merged, according to their
overlap sequence. Operational taxonomic units (OTUs) were clustered with a 97% similarity cutoff using
UPARSE (v7.1 [http://drive5.com/uparse/]), and chimeric sequences identified and removed using
UCHIME. The taxonomy of each 16S rRNA gene sequence was analyzed using the RDP Classifier algo-
rithm (http://rdp.cme.msu.edu/) against the Silva (SSU128) 16S rRNA database, with a 70% confidence
threshold. Sample biodiversity was calculated using the ACE, Chao1, and Shannon indices by applying a
Wilcoxon rank sum test. Beta-diversity measures dependent on Bray-Curtis and unweighted-UniFrac dis-
tance values were calculated using mothur. LEfSe analysis was conducted to identify bacterial taxa dif-
ferentially represented between different groups at the phylum to genus taxonomy level (biomarkers)
by one-against-all (less strict). To determine the effect of microbiota interacting with Apparent perform-
ance, redundancy analysis (RDA) was performed at the genus level using the R language vegan packet
on Spearman correlation analysis (RDA 2014).
Statistical analysis. Statistical analyses were performed using SPSS 20.0 software (IBM, New York,
NY). Data were evaluated by one-way ANOVA, and the differences between means assessed using
Duncan’s test. A P value of ,0.05 was considered statistically significant. The data were evaluated by
Spearman correlation analysis of the Euclidean distance.
Data availability. Raw reads were deposited into the NCBI Sequence Read Archive database under
accession number SRP268238.
SUPPLEMENTAL MATERIAL
Supplemental material is available online only.
FIG S1, JPG file, 0.9 MB.
TABLE S1, DOCX file, 0.03 MB.
ACKNOWLEDGMENTS
Financial support for this research was provided by the Earmarked Fund for
Modern Agro-industry Technology Research System of China (CARS-34) and the
Science and Technology Research Key Project of Education Department of Henan
Province (19B230012).
REFERENCES
1. Xiao L, Estelle J, Kiilerich P, Ramayo-Caldas Y, Xia Z, Feng Q, Liang S, 8. Rainone V, Schneider L, Saulle I, Ricci C, Biasin M, Al-Daghri NM, Giani E,
Pedersen AO, Kjeldsen NJ, Liu C, Maguin E, Dore J, Pons N, Le Chatelier E, Zuccotti GV, Clerici M, Trabattoni D. 2016. Upregulation of inflammasome
Prifti E, Li J, Jia H, Liu X, Xu X, Ehrlich SD, Madsen L, Kristiansen K, Rogel- activity and increased gut permeability are associated with obesity in chil-
Gaillard C, Wang J. 2016. A reference gene catalogue of the pig gut micro- dren and adolescents. Int J Obes (Lond) 40:1026–1033. https://doi.org/10
biome. Nat Microbiol 1:16161. https://doi.org/10.1038/nmicrobiol.2016.161. .1038/ijo.2016.26.
2. Heinritz S, Mosenthin R, Weiss E. 2013. Use of pigs as a potential model 9. Shulman RJ, Eakin MN, Czyzewski DI, Jarrett M, Ou CN. 2008. Increased gas-
for research into dietary modulation of the human gut microbiota. Nutr trointestinal permeability and gut inflammation in children with functional
Res Rev 26:191–209. https://doi.org/10.1017/S0954422413000152. abdominal pain and irritable bowel syndrome. J Pediatr 153:646–650.
3. Kim SW, Weaver AC, Shen YB, Zhao Y. 2013. Improving efficiency of sow https://doi.org/10.1016/j.jpeds.2008.04.062.
productivity: nutrition and health. J Anim Sci Biotechnol 4:26. https://doi 10. Moreira APB, Texeira TFS, Ferreira AB, Peluzio MCG, Alfenas RCG. 2012.
.org/10.1186/2049-1891-4-26. Influence of a high-fat diet on gut microbiota, intestinal permeability, and
4. Mor G, Aldo P, Alvero AB. 2017. The unique immunological and microbial metabolic endotoxaemia. Br J Nutr 108:801–809. https://doi.org/10.1017/
aspects of pregnancy. Nat Rev Immunol 17:469–482. https://doi.org/10 S0007114512001213.
.1038/nri.2017.64. 11. Thum C, Cookson A, Otter D, McNabb W, Hodgkinson A, Dyer J, Roy N.
5. Berchieri-Ronchi CB, Kim SW, Zhao Y, Correa CR, Yeum KJ, Ferreira AL. 2011. 2012. Can nutritional modulation of maternal intestinal microbiota influ-
Oxidative stress status of highly prolific sows during gestation and lacta- ence the development of the infant gastrointestinal tract? J Nutr
tion. Animal 5:1774–1779. https://doi.org/10.1017/S1751731111000772. 142:1921–1928. https://doi.org/10.3945/jn.112.166231.
6. Jabbour HN, Sales KJ, Catalano RD, Norman JE. 2009. Inflammatory pathways 12. Tamburini S, Shen N, Wu HC, Clemente JC. 2016. The microbiome in early
in female reproductive health and disease. Reproduction 138:903–919. life: implications for health outcomes. Nat Med 22:713–722. https://doi
https://doi.org/10.1530/REP-09-0247. .org/10.1038/nm.4142.
7. Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. 2012. 13. Mach N, Berri M, Estelle J, Levenez F, Lemonnier G, Denis C, Leplat JJ,
The effects of oxidative stress on female reproduction: a review. Reprod Chevaleyre C, Billon Y, Dore J, Rogel-Gaillard C, Lepage P. 2015. Early-life
Biol Endocrinol 10:49. https://doi.org/10.1186/1477-7827-10-49. establishment of the swine gut microbiome and impact on host
phenotypes. Environ Microbiol Rep 7:554–569. https://doi.org/10.1111/ 32. Ferguson EM, Slevin J, Hunter MG, Edwards SA, Ashworth CJ. 2007. Bene-
1758-2229.12285. ficial effects of a high fibre diet on oocyte maturity and embryo survival
14. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, Kim SG, Li in gilts. Reproduction 133:433–439. https://doi.org/10.1530/REP-06-0018.
H, Gao Z, Mahana D, Zarate Rodriguez JG, Rogers AB, Robine N, Loke P, 33. Li Y, Zhang L, Liu H, Yang Y, He J, Cao M, Yang M, Zhong W, Lin Y, Zhuo Y,
Blaser MJ. 2014. Altering the intestinal microbiota during a critical devel- Fang Z, Che L, Feng B, Xu S, Li J, Zhao X, Jiang X, Wu D. 2019. Effects of
opmental window has lasting metabolic consequences. Cell 158:705–721. the ratio of insoluble fiber to soluble fiber in gestation diets on sow per-
https://doi.org/10.1016/j.cell.2014.05.052. formance and offspring intestinal development. Animals (Basel) 9:411.
15. Koren O, Goodrich JK, Cullender TC, Spor A, Laitinen K, Backhed HK, https://doi.org/10.3390/ani9070422.
Gonzalez A, Werner JJ, Angenent LT, Knight R, Backhed F, Isolauri E, 34. Brambillasca S, Zunino P, Cajarville C. 2015. Addition of inulin, alfalfa and
Salminen S, Ley RE. 2012. Host remodeling of the gut microbiome and citrus pulp in diets for piglets: influence on nutritional and faecal parame-
metabolic changes during pregnancy. Cell 150:470–480. https://doi.org/ ters, intestinal organs, and colonic fermentation and bacterial popula-
10.1016/j.cell.2012.07.008. tions. Livestock Sci 178:243–250. https://doi.org/10.1016/j.livsci.2015.06
16. Sahasrabudhe N, Beukema M, Tian L, Troost B, Scholte J, Bruininx E, .003.
Bruggeman G, van den Berg M, Scheurink A, Schols H, Faas M, de Vos P. 35. Zou J, Chassaing B, Singh V, Pellizzon M, Ricci M, Fythe MD, Kumar MV,
2018. Dietary fiber pectin directly blocks Toll-like receptor 2-1 and pre- Gewirtz AT. 2018. Fiber-mediated nourishment of gut microbiota protects
vents doxorubicin-induced ileitis. Front Immunol 9:383. https://doi.org/ against diet-induced obesity by restoring IL-22-mediated colonic health.
10.3389/fimmu.2018.00383. Cell Host Microbe 23:41–53.e4. https://doi.org/10.1016/j.chom.2017.11
17. Luu M, Monning H, Visekruna A. 2020. Exploring the molecular mecha- .003.
nisms underlying the protective effects of microbial SCFAs on intestinal 36. Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M,
tolerance and food allergy. Front Immunol 11:1225. https://doi.org/10 Pudlo NA, Kitamoto S, Terrapon N, Muller A, Young VB, Henrissat B,
.3389/fimmu.2020.01225. Wilmes P, Stappenbeck TS, Nunez G, Martens EC. 2016. A dietary fiber-
18. Gomez-Gallego C, Garcia-Mantrana I, Salminen S, Collado MC. 2016. The deprived gut microbiota degrades the colonic mucus barrier and enhan-
human milk microbiome and factors influencing its composition and ac- ces pathogen susceptibility. Cell 167:1339–1353. https://doi.org/10.1016/
tivity. Semin Fetal Neonatal Med 21:400–405. https://doi.org/10.1016/j j.cell.2016.10.043.
.siny.2016.05.003. 37. Foye OT, Huang IF, Chiou CC, Walker WA, Shi HN. 2012. Early administra-
19. Nakajima A, Kaga N, Nakanishi Y, Ohno H, Miyamoto J, Kimura I, Hori S, tion of probiotic Lactobacillus acidophilus and/or prebiotic inulin attenu-
Sasaki T, Hiramatsu K, Okumura K, Miyake S, Habu S, Watanabe S. 2017. ates pathogen-mediated intestinal inflammation and Smad 7 cell signal-
Maternal high fiber diet during pregnancy and lactation influences regula- ing. FEMS Immunol Med Microbiol 65:467–480. https://doi.org/10.1111/j
tory T cell differentiation in offspring in mice. J Immunol 199:3516–3524. .1574-695X.2012.00978.x.
https://doi.org/10.4049/jimmunol.1700248. 38. Cheng C, Wei H, Yu H, Xu C, Jiang S, Peng J. 2018. Metabolic syndrome
20. Cheng C, Wei H, Xu C, Xie X, Jiang S, Peng J. 2018. Maternal soluble fiber during perinatal period in sows and the link with gut microbiota and
diet during pregnancy changes the intestinal microbiota, improves metabolites. Front Microbiol 9:1989. https://doi.org/10.3389/fmicb.2018
growth performance, and reduces intestinal permeability in piglets. Appl .01989.
Environ Microbiol 84:e01047-18. https://doi.org/10.1128/AEM.01047-18. 39. Wang J, Feng C, Liu T, Shi M, Wu G, Bazer F. 2017. Physiological alterations
21. Costermans N, Teerds K, Keijer J, Knol E, Koopmanschap R, Kemp B, Soede associated with intrauterine growth restriction in fetal pigs: causes and
N. 2019. Follicular development of sows at weaning in relation to esti- insights for nutritional optimization. Mol Reprod Dev 84:897–904. https://
mated breeding value for within-litter variation in piglet birth weight. doi.org/10.1002/mrd.22842.
Animal 13:554–563. https://doi.org/10.1017/S1751731118001684. 40. Yang J, Qian K, Wang C, Wu Y. 2018. Roles of probiotic lactobacilli inclu-
22. Sawicki CM, Livingston KA, Obin M, Roberts SB, Chung M, McKeown NM. sion in helping piglets establish healthy intestinal inter-environment for
2017. Dietary fiber and the human gut microbiota: application of evi- pathogen defense. Probiotics Antimicrob Proteins 10:243–250. https://
analyses reveal key innate immune signatures in the host response to the metabolite butyrate, suppresses colonic inflammation and carcinogene-
gastrointestinal pathogen Campylobacter concisus. Infect Immun 83:832–845. sis. Immunity 40:128–139. https://doi.org/10.1016/j.immuni.2013.12.007.
https://doi.org/10.1128/IAI.03012-14. 54. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM,
51. Heimesaat M, Fischer A, Plickert R, Wiedemann T, Loddenkemper C, Glickman JN, Garrett WS. 2013. The microbial metabolites, short-chain
Göbel U, Bereswill S, Rieder G. 2014. Helicobacter pylori induced gastric fatty acids, regulate colonic Treg cell homeostasis. Science 341:569–573.
immunopathology is associated with distinct microbiota changes in the https://doi.org/10.1126/science.1241165.
large intestines of long-term infected Mongolian gerbils. PLoS One 9: 55. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H,
e100362. https://doi.org/10.1371/journal.pone.0100362. Cross JR, Pfeffer K, Coffer PJ, Rudensky AY. 2013. Metabolites produced
52. Macia L, Tan J, Vieira AT, Leach K, Stanley D, Luong S, Maruya M, Ian by commensal bacteria promote peripheral regulatory T-cell generation.
McKenzie C, Hijikata A, Wong C, Binge L, Thorburn AN, Chevalier N, Ang Nature 504:451–455. https://doi.org/10.1038/nature12726.
C, Marino E, Robert R, Offermanns S, Teixeira MM, Moore RJ, Flavell RA, 56. National Research Council. 2012. Nutrient requirements of swine, 11th
Fagarasan S, Mackay CR. 2015. Metabolite-sensing receptors GPR43 and ed. National Academies Press, Washington, DC. https://doi.org/10.17226/
GPR109A facilitate dietary fibre-induced gut homeostasis through regula- 13298.
tion of the inflammasome. Nat Commun 6:6734. https://doi.org/10.1038/ 57. Singh KM, Jisha TK, Reddy B, Parmar N, Patel A, Patel AK, Joshi CG. 2015.
ncomms7734. Microbial profiles of liquid and solid fraction associated biomaterial in
53. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, buffalo rumen fed green and dry roughage diets by tagged 16S rRNA
Prasad PD, Manicassamy S, Munn DH, Lee JR, Offermanns S, Ganapathy V. gene pyrosequencing. Mol Biol Rep 42:95–103. https://doi.org/10.1007/
2014. Activation of Gpr109a, receptor for niacin and the commensal s11033-014-3746-9.