ArticlePDF Available

Influence of medium-chain fatty acids and short-chain organic acids on jejunal morphology and intra-epithelial immune cells in weaned piglets

Authors:

Abstract and Figures

Medium-chain fatty acids (MCFA) and short-chain organic acids (SOA) are often used as feed additives in piglet diets. There are limited studies in pigs describing the impact of MCFA or SOA on gut morphology and the local immune system. The aim of this study was to investigate whether the supplementation of SOA (0.41% fumaric acid and 0.32% lactic acid), or the combination of SOA with MCFA (0.15% caprylic and capric acid) would have effects on gut morphology and intestinal immune cells in weaned piglets. A total number of 72 weaned piglets were randomly allocated into three experimental groups. Tissue samples of six animals per group were used to investigate the potential impact of the feed additives on villus length and crypt depth of the jejunum and to quantify intra-epithelial lymphocytes (IEL). CD3-positive IEL were determined via immunohistochemistry (IHC) and flow cytometry (FC), whereas CD2-, CD5-, CD8β-, CD16- and γδ TCR-positive IEL were only analysed by FC. The supplementation of MCFA and SOA did not significantly affect morphometric data. The FC data indicated that SOA significantly increased the quantity of CD2(-) CD8(-) γδ T cells in the jejunum epithelium. Both IHC and FC analyses of pig jejunum confirmed that the majority of IEL expressed the surface marker CD3 and could be classified as cytotoxic T lymphocytes. In conclusion, the data indicated that SOA increased the proportion of CD2(-) CD8(-) γδ T cells in the jejunal epithelium. Thus, SOA might enable a beneficial effect on the local immunity by increasing the constitutive number of potential effector cells to defeat infectious diseases.
Content may be subject to copyright.
ORIGINAL ARTICLE
Influence of medium-chain fatty acids and short-chain organic
acids on jejunal morphology and intra-epithelial immune cells in
weaned piglets
F. Ferrara
1,2
, L. Tedin
1
, R. Pieper
1
, W. Meyer
3
and J. Zentek
1
1 Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universit
at Berlin Berlin, Germany
2 Institute of Vegetative Physiology, Charit
e University Hospital and Center of Cardiovascular Research Berlin, Germany, and
3 Institute of Anatomy, Foundation, University of Veterinary Medicine Hannover, Germany
Summary
Medium-chain fatty acids (MCFA) and short-chain organic acids (SOA) are often used as feed additives in
piglet diets. There are limited studies in pigs describing the impact of MCFA or SOA on gut morphology and
the local immune system. The aim of this study was to investigate whether the supplementation of SOA
(0.41% fumaric acid and 0.32% lactic acid), or the combination of SOA with MCFA (0.15% caprylic and
capric acid) would have effects on gut morphology and intestinal immune cells in weaned piglets. A total
number of 72 weaned piglets were randomly allocated into three experimental groups. Tissue samples of six
animals per group were used to investigate the potential impact of the feed additives on villus length and
crypt depth of the jejunum and to quantify intra-epithelial lymphocytes (IEL). CD3-positive IEL were deter-
mined via immunohistochemistry (IHC) and flow cytometry (FC), whereas CD2, CD5, CD8b, CD16
and cd TCR-positive IEL were only analysed by FC. The supplementation of MCFA and SOA did not signifi-
cantly affect morphometric data. The FC data indicated that SOA significantly increased the quantity of
CD2
CD8
cd T cells in the jejunum epithelium. Both IHC and FC analyses of pig jejunum confirmed that
the majority of IEL expressed the surface marker CD3 and could be classified as cytotoxic T lymphocytes. In
conclusion, the data indicated that SOA increased the proportion of CD2
CD8
cd T cells in the jejunal
epithelium. Thus, SOA might enable a beneficial effect on the local immunity by increasing the constitutive
number of potential effector cells to defeat infectious diseases.
Keywords piglets, small intestinal morphology, local immunity, immunohistochemistry, flow cytometry
Correspondence F. Ferrara, Institute of Vegetative Physiology, Charit
e University Hospital, Hessische Straße 3-4, 10115 Berlin, Germany.
Tel: +49 30 528 447; Fax: + 49 30 528 972; E-mail: fabienne.ferrara@charite.de
Received: 21 April 2015; accepted: 26 January 2016
Introduction
Weaning is a challenge for the piglet including separa-
tion from the mother, environmental and nutritional
changes. This may set the scene for digestive disorders,
reduced performance and even increased mortality
(Campbell et al., 2013). Feed composition is an impor-
tant driving factor for feed intake, energy and nutrient
supply to enterocytes and subsequently affects epithe-
lial structure and integrity (Le Dividich and S
eve,
2000; Lall
es et al., 2004, 2007; Zabielski et al., 2008).
In addition, nutritional factors and changes in the gut
microbial ecosystem may affect the local immune
response in the piglet gut during the weaning period
(Le Dividich and S
eve, 2000; Lall
es et al., 2004, 2007;
Zabielski et al., 2008). Transient changes in intestinal
morphology and function (e.g. reduction of villus
length and enzyme activity) within the first 5 days
following weaning may therefore result from the com-
plex interaction between diet, intestinal microbiota
and the local immune system (Pluske et al., 1997;
Stokes et al., 2004). The mucosa-associated immune
system is an important factor determining the balance
between tolerance against food antigens or autochtho-
nous intestinal bacteria or local defence reactions
against putative pathogens or other antigens. Local
intestinal immune response is mainly driven by com-
plex interactions between innate and acquired mecha-
nisms of the intestinal associated local immune system
(Pabst and Rothk
otter, 1999; Vega-Lopez et al., 2001;
Stokes et al., 2004). Intra-epithelial lymphocytes
(IEL) as part of the gut-associated immune system are
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH 1
DOI: 10.1111/jpn.12490
located between the enterocytes, and in pigs, they are
relatively equally distributed along the villus axis
(Vega-Lopez et al., 2001). Based on the close proxim-
ity to the intestinal lumen, it is hypothesized that IEL
may play an important role within the local immune
defence (Pabst, 1987). Maintaining the intestinal
physiological and intestinal barrier function in wean-
ing piglets is of high importance for the prevention of
digestive disorders.
Medium-chain fatty acids (MCFA) and short-chain
organic acids (SOA) constitute rapidly available
energy sources for intestinal and extra-intestinal tis-
sues and could be useful particularly in feeding
weaned piglets (Lueck, 1980; Borum, 1992; Heo et al.,
2002). MCFA and SOA can be utilized by enterocytes
as energy source and can attenuate negative effects of
weaning on villus length and crypt depth in piglets
(Dierick et al., 2002a,b, 2003; Lee et al., 2007). Addi-
tionally, previous studies suggest that MCFA or SOA
can modulate the number of intra-epithelial lympho-
cytes (IEL), the lymphocyte proliferation rate and
jejunal cytokine expression (Dierick et al., 2002a,b,
2003; Lee et al., 2007; Kuang et al., 2015).
The focus of this study was to investigate the effect
of SOA and the combination of SOA with MCFA on
gut morphology and IEL populations in weaned pig-
lets, with focus on synergistic effects of both feed addi-
tives.
Material and methods
This study was performed in accordance with the Ger-
man ethical and animal care guidelines and approved
by the State Office of Health and Social Affairs ‘Lan-
desamt f
ur Gesundheit und Soziales Berlin’ (LAGeSo)
(registration number G0293/09).
Animals, housing and diets
A total number of 72 castrated piglets (Duroc x Pi
e-
train) (weaned at 25 1 days of age) were used in
three consecutive experimental feeding trials to evalu-
ate the influence of the feed additives on performance
parameters. In each trial, n=24 piglets were ran-
domly allocated into three experimental groups with
eight animals each and two piglets per flat deck pen
resulting in a total of n=72 piglets. Performance data
have been described in a previous publication (Zentek
et al., 2013). The room temperature was 28 °C at the
beginning of the feeding trial and was gradually
decreased to 22 °C within 10 days. The humidity was
65%. A light programme provided a 16-h light and
8-h dark period. Dietary treatments were as follows
(Table 1): a commercial starter diet (CON), the starter
diet containing 1.05% of a SOA (with 39.8% fumaric
acid, 31.% lactic acid and silicium dioxide as a carrier,
SOA), and the starter diet containing SOA and 0.3%
of a MCFA premix (with 50% of caprylic and capric
acid).
Sampling
After a 28-days experimental feeding trail, three pig-
lets per group and per consecutive period, respec-
tively, were euthanized by intracardial application of
a tetracainhydrochloride, mebezoniumiodide and
embutramide (T61
â
; Intervet, Unterschleissheim,
Germany), after a deep anaesthesia with a combina-
tion of ketamine hydrochloride (Ursotamin
â
, 10%;
Serumwerk Bernburg AG, Bernburg, Germany) and
azaperon (Stresnil
â
, Jansen-Cilag, Neuss, Germany)
(1/0.2 mg per kg BM). In total, six animals (two
animals each feeding trail) per group were used to
investigate potential effects of the feed additives on
Table 1 Analysed and calculated nutritional composition of the piglet
diet (as-fed basis)*
Item CON SOA SOA+MCFA
Analysed
Dry matter, g/kg 888.8 887.5 888.6
Crude ash. g/kg 62.9 64.5 67.3
Crude protein, g/kg 207.3 202.6 204.0
Crude fat, g/kg 61.2 59.1 62.3
Crude fibre, g/kg 46.5 49.5 50.3
Ca, g/kg 9.7 9.6 9.8
P, g/kg 5.5 5.5 5.5
Na, g/kg 2.6 2.8 2.9
K, g/kg 8.1 8.1 8.0
Mg, g/kg 1.6 1.6 1.6
Fe, mg/kg 451 445 467
Zn, mg/kg 142 130 128
Cu, mg/kg 114 123 126
Buffering capacity, mEq/kg 545 510 495
pH 4.69 4.41 4.38
L-lactic acid, g/kg 0.45 3.97 3.74
D-lactic acid, g/kg 0.09 0.13 0.11
Fumaric acid, g/kg <0.05 <0.05 <0.05
Caprylic acid, g/kg 0.09 0.08 0.52
Capric acid, g/kg 0.06 0.08 0.52
Calculated
Lys, g/kg 12.5 12.5 12.5
Met, g/kg 4.69 4.69 4.69
Thr, g/kg 8.45 8.45 8.45
Trp, g/kg 2.63 2.63 2.63
ME, MJ/kg 13.8 13.8 13.8
*or +OA, without or with organic acid; or +MCFA, without or with
medium-chain fatty acid.
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH2
MCFA and SOA in piglets F. Ferrara et al.
gut morphology and intra-epithelial cells (IEL). Tissue
samples were taken at approximately 50% of the
entire length of the jejunum avoiding taking tissue
with discrete or parts of the continuous payers patch.
Histological preparation
A first jejunum tissue sample with the length of 5 cm
was removed immediately post-mortem and sliced on
the antimesenterial side, stretched on cork plates and
washed with phosphate-buffered saline (PBS) at room
temperature. The tissue sample was then quickly
transferred (mucosal surface facing downwards) into
the fixation medium 4% neutral buffered formalin
solution (37% formol +29PBS in aqua dest.). In
general, all samples were embedded for histological
examinations according to Hornickel (2009), with
minor modifications, that is during the last step,
incubation of tissue was performed with xylol-I for
30 min (combination of 1:1 xylol and 96% alcohol),
followed by three times washing with pure xylol and
transfer into warm paraffin (60 °C) overnight (12
14 h).
Morphometric analyses
Haematoxylineosin staining was performed by a
standard protocol (Hornickel, 2009). Well-orientated
villus length and crypt depth were measured by an
investigator blinded to outcome with a standard light
microscope (Photomicroscope III; Zeiss, Jena, Ger-
many) with a digital microscope camera (DP 72;
Olympus, Hamburg, Germany)and the related analy-
sis software (Cell Sense Software; Olympus). Morpho-
metric analyses were performed on four slices from
each animal. Villus length was defined as a line
between the villus top and base, whereas crypt depth
was defined as a conjunction line between the open-
ing of the crypt and their base. Per slice, five villi and
crypts were measured.
Immunohistochemical analysis
To detect CD3-positive IELs by immunohistochem-
istry (IHC), the monoclonal antibody PPT3 (Table 2)
was applied to the 5 lm slices after antigen retrieval
within 30 min in citrate buffer (9698 °C). To avoid
non-specific binding of the Fc part of the respective
PPT3 primary antibody, an isotype control was con-
ducted with a non-specific antibody (DAKO, Golstrup,
Denmark), at which the isotype control was applied in
the same protein concentration as the primary anti-
body (Fig. 1). Both antibodies were diluted with 1%
bovine serum albumin (BSA) and PBS. In summary,
an indirect IHC method was performed. To visualize
the primary antibody, a two-step indirect method was
used by the EnVision
â
mouse system (EnVision+Sys-
tem-HRP, mouse K4007, Dako, Golstrup, Denmark)
whereas the secondary antibody was conjugated with
a horseradish peroxidase (HRP)-labelled dextran poly-
mer (goat anti-mouse IgG
1
). In general, the IHC pro-
tocol was performed according to a general procedure
which was published previously (Hornickel, 2009).
For the evaluation of the counting accuracy, a double-
blinded quantification of CD3-positive IEL was per-
formed only on complete and intact villi (four slices
per animal, five villi per slice) and expressed among
100 enterocytes.
Flow cytometric analysis
For the later phenotypic analysis of the IELs via flow
cytometry (FC), an additional 20 cm of the small intes-
tine was taken, adjacent to the jejunal sample that was
chosen for the IHC. The monoclonal primary and fluo-
rescence-labelled secondary antibodies used to identify
IEL populations are listed in Tables 2 and 3.
Cell isolation
The isolation of the cells from the middle of jejunum
tissue sample was performed as described previously
(Mafamane et al., 2011). Briefly: tissues were imme-
diately transferred into fresh PBS solution and
everted to expose the intestinal mucosa. Following
that, tissues were incubated in Hanks0balanced salt
solution (HBSS)-DTT (dithiothreitol) medium and
(5 min and 37 °C) and HBSS-EDTA medium. Cell
suspensions were filtered through a sterile synthetic
gauze (210-mm nylon mesh) and centrifugated at
600 gfor 10 min at 4 °C. Cell pellets were resus-
pended with approximately 10 ml of Roswell Park
Memorial Institute Medium No. 1640 (RPMI) med-
ium and incubated with 50 ll DNase for 5 min at
37 °C and centrifuged (same conditions as described
above). The obtained cell pellet was mixed with Per-
coll (25%) solution and centrifuged (600 g, 20 min).
Finally, the cell pellet was resuspended with RPMI
medium and centrifuged (300 g,4°C, for 10 min)
and suspended with lysis buffer (ammoniumchloride
solution).
Flow cytometry
The staining of the cell preparation for the unla-
belled primary antibodies CD3, CD2, CD8band
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH 3
F. Ferrara et al. MCFA and SOA in piglets
Po-TcR1-N4 (Table 2) was performed as previously
published (Mafamane et al., 2011). For every reac-
tion, 1 910
6
cells were solved to saturating concen-
trations of the antibody in a volume of 100 llof
PBS and 0.2% BSA for 20 min on ice. Cells were
incubated with the fluorescence-labelled antibody
(Table 3). Finally, cells were analysed by the
FACSCalibur flow cytometer (Becton Dickinson Bio-
science, San Jos
e, CA, USA), using the computer
software CellQuest pro
â
(Becton Dickinson Bio-
science), by evaluating the relative distribution of
the diverse IEL phenotypes in the middle of jeju-
num. Per cent values were calculated within the
lymphocyte gate (=100%).
Additionally, the relation of IEL phenotypes among
100 enterocytes was calculated using the counted
number of CD3-positive IEL using the following calcu-
lation formula:
Number of CD3þcells=100enterocytes
ð% of population measured per FC=% of CD3þIEL in FCÞ
Statistical analyses
The statistical evaluation was performed by SPSS 19.0
(SPSS Inc., Chicago IL, USA). The normal distribution
of the data was tested with the KolmogorovSmirnov
test including Lilliefors test. Normally distributed data
were expressed as mean and standard deviation; non-
normally distributed data are expressed as median,
minimum and maximum (villus length and crypt
depth). Single variance analyses were performed on
normally distributed data, followed by the LSD post
hoc test in case of p <0.05. The KruskalWallis test
was performed on non-normally distributed data
followed by WilcoxonMannWhitney test in case of
(a) (b)
(c) (d)
Fig. 1 Brown-stained CD3-positive IEL in the mid-jejunum (control group); LM, 9400; (a, b) isotype control of primary antibody PPT3 without (a) and
with counter staining (b); (c, d) antigen retrieval with citrate buffer (30 min, 9698 °C) without (c) and with counter staining (d).
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH4
MCFA and SOA in piglets F. Ferrara et al.
significance to determine the differences between
groups. A p-value of 0.05 was considered significant
for all data, whereas a p-value of 0.1 was considered
as tendency. p-values which are listed in all tables
based on a single variance analyses (normally dis-
tributed data) or KruskalWallis test (non-normally
distributed data).
Results
Histological and Immunohistological measurements
Villus length and crypt depth in the mid-jejunum
were not significantly influenced by dietary treat-
ment. However, a trend towards longer villi was
determined with the SOA+MCFA group as compared
to the control (Table 4).
Table 4 Immunohistochemical counts of CD3
+
-
IEL and villus length and crypt depth in the mid-
dle of jejunum
Group
CON SOA SOA+MCFA p-Value
CD3
+
-IEL*60.4 6.6 61.5 4.7 58.7 7.7 0.549
Villus length400 (230500) 410 (300530) 420 (260540) 0.098
Crypt depth130 (70260) 140 (60300) 140 (80290) 0.600
CON, control; SOA, short-chain organic acids (fumaric and lactic acid); MCFA, medium-chain fatty
acids (caprylic acid and capric acid); CD, cluster of differentiation; IEL, intra-epithelial lymphocytes;
p, probability value from an analysis of variance (CD3 measurement) or KruskalWallis test (villus
length and crypt depth).
*Mean of CD3-positive IELs determined from individual data from both microscopy counts,
expressed among 100 enterocytes; results of measured mid-jejunum.
Villus length (lm).
Crypt depth (lm): median (minimummaximum).
Table 2 Primary antibodies to differentiate IEL
in the small intestine in piglets Primary
Antibody Isotype Specificity Dilution Method Vendor
PPT3* Mouse
IgG
1
K
Porcine CD3e1:100
1:200
IHC
FC
SouthernBiotech.,
Birmingham,
AL, USA
Anti-mouse
IgG
1
Mouse
IgG
1k
Non 1:100 IHC Dako, Golstrup,
Denmark
CD2 Mouse
IgG
2a
Porcine
CD2
1:200 FC VMRD Inc.
Pullman,
WA, USA
PGBL22AMouse
IgG
1
Porcine
TcR1-N4
1:100 FC VMRD Inc.
PG164A Mouse
IgG
2a
Porcine CD8b1:100 FC VMRD Inc.
G7 (CD16) Mouse
IgG
1
Porcine CD16 1:100 FC Acris Antibodies
Herford Germany
CD5 Mouse
IgG
1
Porcine CD5 1:100 FC VMRD Inc.
IHC, immunohistochemistry; FC, flow cytometry.
*Primary antibody was used for IHC and FC.
Isotype control.
Primary antibody for the detection of cd
+
T cells, binds on d-chain; all listed antibodies are mono-
clonal.
Table 3 Secondary antibodies to differentiate IEL by FC
Secondary antibody* Primary antibody Dilution Stain
Goat Anti-mice PPT3 1:100 FITC
IgG
1
PGBL22A 1:200 PE
G7 (CD16) 1:200 PE
CD5 1:200 PE
Goat Anti-mice CD2 1:100 FITC
IgG
2a
CD8b1:100 FITC
FITC, fluoresceine isothiocyanate; PE, phycoerythrin.
*All secondary antibodies were produced by SouthernBiotech.
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH 5
F. Ferrara et al. MCFA and SOA in piglets
Similarly, no significant differences for CD3-positive
IELs were observed by IHC in jejunal tissue samples
(Table 4).
Characterization of IELs by flow cytometry
The majority of IEL expressed the surface marker CD3
(Tables 5 and 6). The majority of CD3-positive cells
could be classified based on the data obtained by FC as
cytotoxic T lymphocytes (CD3
+
CD8
+ab
). There were
no significant differences in CD3 cell count between
CON and the treatment groups or within the
treatment groups (Tables 5 and 6). Additionally, a low
number of (CD2
CD8
)cd T cells could be detected
by FC (Tables 5 and 6). The data indicate a significant
(p =0.027) increase of the quantity of (CD2
CD8
)
cd T cells in the epithelium in the jejunum in the
piglets from group SOA.
Discussion
The hypothesis of this study was that SOA and MCFA
would affect gut morphology and the local intestinal
immune system in weaned piglets due to the effects
on the intestinal microbiota and their role as energy
source for the epithelial cells. Animal performance
data and changes in gastrointestinal microbial ecology
have been described previously (Zentek et al., 2013).
Structural and functional changes of the small intes-
tine have been often characterized by a reduction of
villus length and increased crypt depth and enzyme
activity, or higher intestinal permeability, thus indi-
cating disturbed mucosa integrity during the first 3
5 days post-weaning (Boudry et al., 2004; Montagne
et al., 2007; Smith et al., 2010). This has been shown
to predispose piglets to gastrointestinal diseases and is
associated with lower growth rate post-weaning
(Pluske et al., 1997; Zabielski et al., 2008). Weaning
resulted in a decrease of villus length in the jejunum
from approximate 740390 lm (3 days post-weaning,
15 days of age), whereas the crypt depth increased
from ~180 lmupto210lm (Tang et al., 1999). The
regeneration stage, from days 515 post-weaning, is
characterized by the progressive reconstruction of
physiological structures and digestive capacity (Tang
et al., 1999; Spreeuwenberg et al., 2001; Montagne
et al., 2007). Within the first 34 days of age, villus
length increased to 400500 lm (Tang et al., 1999).
Generally, the obtained data regarding villus length in
this study are in good agreement with previous studies
in weaned piglets (Tang et al., 1999; Montagne et al.,
2007; Martin et al., 2012), at which villus length was
in a range between 400420 lm and crypt depth
between 130140 lmat53(1) days of age. How-
ever, the assumption that dietary supply with SOA or
SOA+MCFA would affect jejunal morphology could
be confirmed only in part because no significant dif-
ferences between the dietary treatments were deter-
mined. The slightly longer villi without differences in
crypt depth in the SOA+MCFA group may indicate a
reduced rate of apoptosis at the tops of the villi. Simi-
larly, the administration of medium-chain triglyc-
erides from Cuphea seed (C. lancelota und C. ignea)in
combination with lipase in the diet of weaned piglets
has been shown to lead to a significant increase in vil-
lus length at the distal small intestine and a decrease
of depth of the crypts (Dierick et al., 2003). In con-
trast, the administration of formic acid resulted in
Table 5 Phenotypes of IEL analysed by FC in the mid-jejunum of the pig-
lets (data as % of total epithelial lymphocytes; mean and standard devia-
tion)
Group
CON SOA SOA+MCFA p-Value
CD3
+
72.0 8.7 82.2 6.9 77.9 8.1 0.117
CD8b
+
46.34 8.3 54.0 8.4 51.5 13.2 0.318
CD2
+
/CD5
33.3 9.6 23.2 9.2 27.6 7.6 0.081
CD16
+
28.7 8.9 27.5 6.9 28.4 5.9 0.905
CD2
+
/cd
+
22.6 4.4 20.7 5.5 19.0 6.3 0.440
CD2
/cd
+
0.6 0.3
b
1.1 0.5
a
0.6 0.3
b
0.027
CON, control group; SOA, short-chain organic acids (fumaric and lactic
acid); MCFA, medium-chain fatty acids (caprylic acid and capric acid);
IEL, intra-epithelial lymphocytes; CD, cluster of differentiation; FC, flow
cytometry; p, probability value from analysis of variance.
a,b
Different indices within a line shows significant differences (p 0.05).
Table 6 The relation of IEL phenotypes among 100 enterocytes, calcu-
lated from the counted number of CD3-positive IEL and FC data (mean
and standard deviation)
Group
CON SOA SOA+MCFA p
CD3
+
-IEL*60.4 6.3 60.3 5.7 58.0 8.0 0.867
CD8b
+
-IEL 36.2 7.8 41.4 5.1 42.3 6.8 0.434
CD2
+
/CD5
-IEL 28.0 7.6 15.4 9.0 20.3 9.5 0.211
CD16
+
-IEL 25.0 4.9 17.8 6.7 20.70 7.6 0.377
CD2
+
/cd
+
-IEL 17.8 3.7 12.4 4.4 11.4 5.6 0.176
CD2
/cd
+
-IEL 0.50 0.3 0.70 0.3 0.4 0.2 0.406
CON, control group; SOA, short-chain organic acids (fumaric and lactic
acid); MCFA, medium-chain fatty acids (caprylic acid and capric acid););
IEL, intra-epithelial Lymphocytes; CD, cluster of differentiation; FC, flow
cytometry;IHC, immunohistochemistry; p, probability value from analy-
sis of variance.
*Mean of CD3-positive IELs determined from combining individual data
from both microscopy counts, expressed per 100 enterocytes.
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH6
MCFA and SOA in piglets F. Ferrara et al.
shorter jejunal villi of early weaned piglets (Man-
zanilla et al., 2004). Consequently, our findings sug-
gest that there might be synergistic positive effects of
combined feeding with MCFA+SOA in weaned pig-
lets. The performance of the piglets was not affected
by MCFA or SOA (Zentek et al., 2013), thus indicat-
ing a more related effect of MCFA directly towards
enterocytes. Consequently, the mechanisms and pos-
sible triggers driving small intestinal cell proliferation
and apoptosis under the influence of SOA and MCFA
are of interest and require further investigations.
In addition, epithelial changes in response to SOA
and MCFA during the early post-weaning period
might be even more important to assess small intesti-
nal functionality during this time.
For the assessment of the impact of both additives
on mucosal immune function, morphometric mea-
surements were combined with quantification of IELs
by IHC and FC. The identical PPT3 antibody was used
to allow the evaluation of results from IHC and FC in
two adjacent intestinal segments. In general, the
number of CD3-positive IELs was not significantly dif-
ferent between the treatment groups. Obviously, the
percentage of CD3-positive IELs analysed by FC was
higher than determined with IHC. This could be either
due to methodological effects, for example different
thicknesses of paraffin blocks or counting procedures
within the IHC method, or the loss of epithelial cells
during the tissue preparation for FC analyses. The
later was emphasized by the decreased total number
of calculated IEL by FC among 100 enterocytes based
on labelled CD3
+
IELs by IHC. However, false-positive
results of IHC were excluded by isotype control anti-
body of the primary antibody, and additionally, to
evaluate cell counting accuracy, a double-blinded trial
was performed. Finally, based on our implementation
of FC preparation, it is not possible to recommend one
of these used methods, for that purpose, it is necessary
to use a validate step, for example histological exami-
nation of the jejunal tissue sample of the number of
left epithelial cells following cell isolation.
There are limited studies in pigs describing the
mode of action of MCFA or SOA on IEL populations
in piglets. A significant decrease in the number of IEL
in the small intestine was observed in piglets fed
Cuphea seeds (Dierick et al., 2003). In the present
study, the number of CD3
+
IELs among 100 entero-
cytes did not differ between treatment groups. In the
literature, there is a controversal discussion regarding
the impact of IEL number as an indicator of mucosal
integrity. A reduced IEL number combined with
longer villi and decreased crypt depth, based on
reduced apoptotic rate, was considered as protective
effect of MCFA (Dierick et al., 2003). On the other
hand, a lower number of intestinal IEL may indicate a
downregulation of local immune responses, due to
the reduced number of effector cells (Vega-Lopez
et al., 1995; Rothk
otter et al., 1999; Gu et al., 2002).
In fact, additional investigations could be useful for
further descriptions of potential effects of MCFA and
SOA on the intestinal local immunity, as indicated by
Kuang et al. (2015), at which both additives downreg-
ulated systemic and jejunal pro-inflammatory cyto-
kine expression (TNF-a) and stimulated the tissue
expression of the regulatory cytokine TGF-b.
In the adult pig, it has been shown that over 50% of
the total T cells in the small intestine were located at
the epithelium (Vega-Lopez et al., 2001). The highest
proportion of IELs is located close to the basement
membrane (53%), whereas most of these cells bear
the CD2
+
CD4
CD8
+
phenotype (Vega-Lopez et al.,
2001). A limited number of double-negative
CD2
+
CD4
CD8
cells are located on the same level of
the enterocyte cell nucleus (Vega-Lopez et al., 1993,
2001; Rothk
otter et al., 1994). CD4
+
, CD16
+
and
TcR1
+
cd cells have been found at low density in the
epithelium (Vega-Lopez et al., 1993; Solano-Aguilar
et al., 2001). In general, T lymphocytes of the pigs can
be differentiated by their T-cell receptor into ab T cells
and cd T cells. The first group can be furthermore
divided into four different subtypes (Yang and Park-
house, 1996; Charerntantanakul and Roth, 2007):
native T-helper cells (CD4
+
CD8
cells), T memory
cells (CD4
+
CD8
+aalow
) and cytotoxic T cells
(CD4
CD8
+ablow
and CD4
CD8
+abhigh
cells). CD8
+
cells are a predominant T-cell subset in antiviral
responses, whereas CD4
+
T cells predominantly occur
during bacterial and parasitic infections (Charerntan-
tanakul and Roth, 2007).
To achieve a wide differentiation of IELs, an addi-
tional jejunum tissue sample was further investigated
by FC. The FC data demonstrated that the majority of
IELs expressed the surface marker CD3. Furthermore,
CD3 and CD5 are not expressed at the surface of natu-
ral killer (NK) cells and may be useful to separate
CD8-positive cd T lymphocytes from NK cells
(Saalm
uller et al., 1994). In summary, the FC data
indicated that SOA significantly increased (p =0.027)
the quantity of CD2
CD8
cd T cells in the epithelium
in the jejunum. Phenotypically, the total porcine cd T
cells are divided into three subsets based on the
surface expression of CD2 and CD8
aa
(Wen et al.,
2012). The non-classical group of cd T cells
(CD2
CD4
CD8
) represents the main proportion of
T cells in young pigs (Yang and Parkhouse, 1996;
Stepanova and Sinkora, 2013) with 1526% (4 weeks
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH 7
F. Ferrara et al. MCFA and SOA in piglets
of age) and 2439% (4 month old piglets) (Yang and
Parkhouse, 1996). The number of non-classical cd T
cells in the blood decrease with the age of 8 months
(Yang and Parkhouse, 1996). All functions of
non-classical cd T cells are until now not fully
understood (Charerntantanakul and Roth, 2007;
Stepanova and Sinkora, 2013). However, the high
number of non-classical group of cd T cells is in
contrast to humans and mice, thus indicating a por-
cine specific lineage (Stepanova and Sinkora, 2013).
In fact, a fraction of non-classical cd T cells has the
ability to downregulate TCRcd within hours in
spleen and thymus and additionally to upregulate
their receptor only in thymus (Stepanova and Sin-
kora, 2013). The proliferation rate of non-classical T
cells in spleen is very low, compared to those of
classical cd T cells (CD2
+
CD4
CD8
,
CD2
+
CD4
CD8
+aa
) (Stepanova and Sinkora, 2013).
Equally to the CD2
+
CD4
CD8
cd T subset (classical
cd T cells), the non-classical cd T cells have mainly
pro-inflammatory function as visible by directly
secreting of IFN-cor by promoting CD4
+
ab T-cell
proliferation (Wen et al., 2012). In contrast to the
examinations of Stepanova and Sinkora (2013), the
study of Wen et al. (2012) revealed that the CD8
subsets can differentiate into CD8
+
by acquiring
CD8 expression in ileum and blood following exper-
imental virus infection, at which the frequencies of
the non-classical cd T cells were reduced in these
compartments. According to the later study, our
results may be indicating a beneficial effect of SOA
by increasing a constitutive higher number of non-
classical T cells to defend pathogen infection, which
appears to be an additional beneficial effect within
the context of prevention of post-weaning (Zentek
et al., 2013).
The cellular processes of antigen identification by cd
T cells appear to be assumed: Unlike ab T cells, these
cells are known to recognize foreign antigens in a
non-MHC-restricted manner directly by their receptor
or like unspecific immune cells with the aid of patho-
gen-associated patterns (PAMPs), by Toll-like recep-
tors, or scavenger receptors (Hedges et al., 2005;
Charerntantanakul and Roth, 2007; Wen et al.,
2012). However, the results of our study indicated
that cytotoxic ab T cells, cd T cells and NK cells repre-
sent a large proportion of IEL; this may indicate an
important role of IEL in local gut immunity, at which
they play important roles in antigen recognition and
immune response (Wen et al., 2012).
Conclusion
In conclusion, the supplementation of SOA or SOA
+MCFA to a starter diet for piglets did not affect sig-
nificantly jejunal morphology. However, a tendency
towards higher villi without changes in crypt depth
may indicate some effects on epithelial cell turnover,
which requires further investigation. Using IHC and
FC revealed that the majority of IEL expressed the
surface marker CD3 and could be classified as cyto-
toxic T lymphocytes without significant differences
between treatments. In summary, feeding SOA
increased the quantity of non-classical cd T cells
which may confer protection against putative patho-
gens or antigens through a higher abundance of
effector cells.
Acknowledgement
The authors wish to thank Mrs. Petra Huck for her
technical assistance with flow cytometry analyses,
Mrs. Karin Briest-Forch for her technical assistance
with histological preparations and Damian McLeod
for help in preparing the manuscript.
References
Borum, P. R., 1992: Medium-chain
triglycerides in formula for preterm neo-
nates: implications for hepatic and
extrahepatic metabolism. Journal of Pedi-
atrics 120, 139145.
Boudry, G.; P
eron, V.; Hu
erou- Luron,
I.; Lall
es, J. P.; S
eve, B., 2004:
Weaning induces both transient and
long- lasting modifications of absorp-
tive, and barrier properties of piglet
intestine. Journal of Nutrition 134,
22562262.
Campbell, J. M.; Crenshaw, J. D.; Polo, J.,
2013: The biological stress of early
weaned piglets. Journal of Animal Science
and Biotechnology 4,14.
Charerntantanakul, W.; Roth, J. A., 2007:
Biology of porcine T lymphocytes.
Animal Health Research Reviews 7,
8192.
Dierick, N. A.; Decuypere, J. A.; Molly,
K.; van Beek, E.; Vanderbeke, E.,
2002a: The combined use of triacyl-
glycerols containing medium-chain
fatty acids (MCFAs) and exogenous
lipolytic enzymes as an alternative for
nutritional antibiotics in piglet nutri-
tion I. In vitro screening of the release
of MCFAs from selected fat sources by
selected exogenous lipolytic enzymes
under simulated pig gastric conditions
and their effects on the gut flora of
piglets. Livestock Production Science 75,
129142.
Dierick, N. A.; Decuypere, J. A.; Molly, K.;
van Beek, E.; Vanderbeke, E., 2002b: The
combined use of triacylglycerols (TAGs)
containing medium chain fatty acids
(MCFAs) and exogenous lipolytic enzymes
as an alternative to nutritional antibiotics
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH8
MCFA and SOA in piglets F. Ferrara et al.
in piglet nutrition. II. In vivo release of
MCFAs in gastric cannulated and slaugh-
tered piglets by endogenous and exoge-
nous lipases; effects on the luminal gut
flora and growth performance. Livestock
Production Science 76,116.
Dierick, N. A.; Decuypere, J. A.; Degeyter,
I., 2003: The combined use of whole
Cuphea seeds containing medium chain
fatty acids and an exogenous lipase in
piglet nutrition. Archiv f
ur Tieren
ahrung
57,4963.
Gu, X.; Li, D.; She, R., 2002: Effect of
weaning on small intestinal structure
and function in the piglet. Archives of
Animal Nutrition 56, 275286.
Hedges, J. F.; Lubick, K. J.; Jutila, M. A.,
2005: cd T cells respond directly to
pathogen-associated molecular patterns.
Journal of Immunology 174, 60456053.
Heo, K. N.; Lin, X.; Han, I. K.; Odle, J.,
2002: Medium-chain fatty acids but not
L-carnitine accelerate the kinetics of
14C triacylglycerol utilization by colos-
trum-deprived newborn pigs. Journal of
Nutrition 132, 19891994.
Hornickel, N., 2009: Investigations on the
innate immunity of the esophagus
epithelium of domesticated mammals.
Thesis, Tier
arztliche Hochschule Han-
nover, Hannover
Kuang, Y.; Wang, Y.; Zhang, Y.; Song, Y.;
Zhang, X.; Lin, Y.; Che, L.; Xu, S.; Wu,
D.; Xue, B.; Fang, Z., 2015: Effects of
dietary combinations of organic acids
and medium chain fatty acids as a
replacement of zinc oxide on growth,
digestibility and immunity of weaned
pigs. Animal Feeding Science and Technol-
ogy 208, 145157.
Lall
es, J. P.; Boudry, G.; Favier, C.; Le Floc
̓
h, N.; Luron, I.; Montagne, L.; Oswald,
I.; Pie
́, S.; Piel, C.; Sève, B., 2004: Gut
function and dysfunction in young pigs.
Animal Research 53, 301316.
Lall
es, J. P.; Bosi, P.; Smidt, H.; Stokes, C.
R., 2007: Nutritional management of
gut health in pigs around weaning. Pro-
ceeding of the Nutrition Society 66, 260
268.
Le Dividich, J.; S
eve, B., 2000: Effects of
underfeeding during the weaning per-
iod on growth, metabolism, and hor-
monal adjustments in the piglet.
Domestic Animal Endocrinology 19,6374.
Lee, D. N.; Liu, R.; Chen, Y. T.; Wang, R.
C.; Lin, S. Y.; Weng, C. F., 2007: Effects
of diets supplemented with organic acids
and nucleotides on growth, immune
response and digestive tract develop-
ment in weaned pigs. Journal of Animal
Physiology and Animal Nutrition 91, 508
518.
Lueck, E., 1980: Antimicrobial Food Addi-
tives. Springer-Verlag, Berlin Heidelberg,
New York.
Mafamane, H.; Szabo, I.; Schmidt, F. G.;
Filter, M.; Walk, N.; Tedin, K.; Scharek-
Tedin, L., 2011: Studies on the effect of
an Enterococcus faecium probiotic on T cell
populations in peripheral blood and
intestinal epithelium and on the suscep-
tibility to Salmonella during a challenge
infection with Salmonella Typhimurium
in piglets. Archives of Animal Nutrition 65,
415430.
Manzanilla, E. G.; Perez, J. F.; Martin, M.;
Kamel, C.; Baucells, F.; Gasa, J., 2004:
Effect of plant extracts and formic acid
on the intestinal equilibrium of early-
weaned pigs. Journal of Animal Science
82, 32103218.
Martin, L.; Pieper, R.; Kr
oger, S.; Goodarzi
Boroojeni, F.; Vahjen, W.; Neumann,
K.; Van Kessel, A. G.; Zentek, J., 2012:
Influence of age and Enterococcus fae-
cium NCIMB 10415 on development of
small intestinal digestive physiology in
piglets. Animal Feed Science and Technol-
ogy 175,6575.
Montagne, L.; Boudry, G.; Favier, C.; Le
Hu
erou- Luron, I.; Lall
es, J. P., 2007:
Main intestinal markers associated with
the changes in gut architecture and
function in piglets after weaning. British
Journal of Nutrition 97,4557.
Pabst, R., 1987: The anatomical basis for
the immune function of the gut. Anat-
omy and Embryology 176, 135144.
Pabst, R.; Rothk
otter, H. J., 1999: Postna-
tal development of lymphocyte subsets
in different compartments of the small
intestine of piglets. Veterinary Immunol-
ogy and Immunopathology 72, 167173.
Pluske, J. R.; Hampson, D. J.; Williams, I.
H., 1997: Factors influencing the struc-
ture and function of the small intestine
in the weaned pig: a review. Livestock
Production Science 51, 215236.
Rothk
otter, H. J.; Kirchhoff, T.; Pabst, R.,
1994: Lymphoid and nonlymphoid cells
in the epithelium and Lamina propria of
the intestinal mucosa in pigs. Gut 35,
15821589.
Rothk
otter, H. J.; M
ollhof, S.; Pabst, R.,
1999: The influence of age and breeding
conditions on the number and prolifera-
tion of intraepithelial lymphocytes in
pigs. Scandinavian Journal of Immunology
50,3138.
Saalm
uller, A.; Hirt, W.; Maurer, S.;
Weiland, E., 1994: Discrimination
between two subsets of porcine CD8
+
cytolytic T lymphoytes by the expres-
sion of CD5 antigen. Immunology 81,
578583.
Smith, F.; Clark, J. E.; Overman, B. L.;
Tozel, C. C.; Huang, J. H.; Rivier, J. E.;
Blikslager, A. T.; Moeser, A. J., 2010:
Early weaning stress impairs develop-
ment of mucosal barrier function in the
porcine intestine. American Journal of
Physiology Gastrointestinal and Liver Physi-
ology 298, 352363.
Solano-Aguilar, G. I.; Vengroski, K. G.;
Beshah, E.; Douglass, L. W.; Lunney, J.
K., 2001: Characterization of lympho-
cyte subsets from mucosal tissue in
neonatal swine. Developmental & Compar-
ative Immunology 25, 245263.
Spreeuwenberg, M. A. M.; Verdonk, J. M.
A. J.; Gaskins, H. R.; Verstegen, M. W.
A., 2001: Small intestine epithelial bar-
rier function is compromised in pigs
with low feed intake at weaning. Journal
of Nutrition 131, 15201527.
Stepanova, K.; Sinkora, M., 2013: Porcine
cd T Lympohocytes can be categorized
into two functionally and developmen-
tally distinct Subsets according to
expression of CD2 and level of TCR. The
Journal of Immunology 190, 21112120.
Stokes, C. R.; Bailey, M.; Haverson, K.;
Harris, C.; Jones, P.; Inman, C.; Pie, S.;
Oswald, I. P.; Williams, B. A.; Akker-
mans, A. D. L.; Sowa, E.; Rothk
otter, H.
J.; Miller, B. G., 2004: Postnatal devel-
opment of the intestinal immune system
in piglets: implications for the process of
the weaning. Animal Research 53, 325
334.
Tang, M.; Laarveld, B.; Van Kessel, A. G.;
Hamilton, D. L.; Estrada, A.; Patience, J.
F., 1999: Effect of segregated early
weaning on postweaning small intesti-
nal development in pigs. Journal of Ani-
mal Science 77, 31913200.
Vega-Lopez, M. A.; Bailey, M.; Stevens,
K.; Stokes, C. R., 1993: Immune cell dis-
tribution in the small intestine of the
pig: immunhistological evidence for an
organized compartmentalization in the
lamina propria. Veterinary Immunology
and Immunopathology 37,4960.
Vega-Lopez, M. A.; Bailey, M.; Telemo, E.;
Stokes, C. R., 1995: Effect of early
weaning on the development of
immune cells in the pig small intestine.
Veterinary Immunology and Immunopathol-
ogy 44, 319327.
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH 9
F. Ferrara et al. MCFA and SOA in piglets
Vega-Lopez, M. A.; Arenas-Contreras, G.;
Bailey, M.; Gonzales-Pozos, S.; Stokes,
C. R.; Ortega, M. G.; Mondragon-Flores,
R., 2001: Development of intraepithelial
cells in the porcine small intestine.
Developmental & Comparative Immunology
8, 147158.
Wen, K.; Bui, T.; Li, G.; Liu, F.; Li, Y.;
Kocher, J.; Yuan, L., 2012: Characteriza-
tion of immune modulating functions of
cd T cell subsets in a gnotobiotic pig
model of human rotavirus infection.
Comparative Immunology, Microbiology and
Infectious Diseases 35, 289301.
Yang, H.; Parkhouse, R. M., 1996:
Phenotypic classification of porcine
lymphocyte subpopulations in blood
and lymphoid tissues. Immunology 89,
4552.
Zabielski, R.; Godlewski, M. M.; Guil-
loteau, P., 2008: Control of develop-
ment of gastrointestinal system in
neonates. Journal of Physiology and Phar-
macology 59(Suppl. 1), 3554.
Zentek, J.; Ferrara, F.; Pieper, R.; Tedin,
L.; Meyer, W.; Vahjen, W., 2013:
Effects of dietary combinations of
organic acids and medium chain fatty
acids on the gastrointestinal microbial
ecology and bacterial metabolites in
the digestive tract of weaning piglets.
Journal of Animal Science 93,
32003210.
Journal of Animal Physiology and Animal Nutrition ©2016 Blackwell Verlag GmbH10
MCFA and SOA in piglets F. Ferrara et al.
... The height of the villi and the depth of the crypts represent the capacity for nutrient absorption and the rate of cell formation and renewal, respectively 40 . The higher the villi, the more epithelial cells and lymphocytes are present, improving the immune response 41,42 . The greater depth of the crypts refers to a higher speed of cell renewal of the villi due to the increase in the rate of epithelial desquamation or inflammatory damage to the intestinal mucosa, resulting in an increase in immature cells with lower secretory, digestive, and absorptive capacity 3,43 . ...
... Like butyric acid monoglyceride, capric and caprylic acid monoglycerides have immunomodulatory activities, negatively regulating the expression of pro-inflammatory cytokines (e.g. interferon gamma) and helping to control an increased response 42,64 . ...
Article
Full-text available
The study aimed to evaluate the effects of supplementing monoglyceride blend in diets without growth promoters on performance, diarrhea occurrence, blood profile, intestinal morphology and pH, mRNA expression of nutrient transporters, inflammatory markers, antioxidant enzymes, and junction proteins in weaned piglets. Forty piglets were randomly allocated to five groups fed the following diets: control (C), or supplemented with 0.75 g/kg of a blend of fatty acids in powder form (PA), or with 3.00 g/kg of a blend of fatty acids in powder form (HPA), or with 0.50 g/kg of a blend of fatty acids in liquid form (LA), or with 2.00 g/kg of a blend of fatty acids in liquid form (HLA). The LA and PA diets reduced (P < 0.05) the occurrence of diarrhea. The pH of intestinal contents was reduced (P < 0.05) in piglets fed monoglycerides blend. Fecal E. coli count tended (0.05 ≤ P < 0.1) to be reduced in piglets receiving all supplemented diets. LA diet increased (P < 0.05) villus height in the duodenum, while others tended to increase it (0.05 ≤ P < 0.1). In the jejunum, all supplemented diets increased (P < 0.05) the goblet cell proportion. In the ileum, PA diet reduced (P < 0.05) crypt depth and increased (P < 0.05) villus: crypt ratio, and PA, HPA, and HLA diets increased (P < 0.05) goblet cell proportion. In the ileum, HPA and LA diets tended to reduce (0.05 ≤ P < 0.1) crypt depth and Peyer’s patch. In the jejunum, LA and HLA diets increased (P < 0.05) the expression of Occludin and HPA increased the expression of Interleukin-10. In conclusion, the supplementation with a monoglyceride blend improves intestinal health and morphology, and local immune response in piglets fed diets without growth promoters.
... Inhibition of Salmonella typhimurium by MCFA has also been shown in an in vitro system of pig cecum 37 . Changes in biological processes have been observed already, for example increased intra-epithelial lymphocytes in jejunum after supplementing piglets with a mixture of MCFAs 38 . The link to immunological processes is more ambiguously in our study; only three pathways could be linked to immunity, i.e., PPAR signaling, innate immune system, and NF-kappa B signaling. ...
Article
Full-text available
Feed additives aiming to improve gastrointestinal health are frequently supplied to piglets after weaning (d28) but might be more effective when administered before weaning. In this period, feed additives can either be administered directly to neonates, or indirectly via sow’s feed. It is yet unknown what the effect of the administration route is on gut functionality and health in the piglets. Therefore, we compared the effect of different dietary interventions on gut functionality after maternal administration (lactation feed) to the neonatal administration route (oral gavage). These feed interventions included medium chain fatty acids (MCFA), beta-glucans (BG), and galacto-oligosaccharides (GOS). For the maternal administration route, MCFA showed a significant difference in alpha diversity parameter, observed species at d1 and one differentially expressed gene (DEG), and 99 DEG at d31. Pathway enrichment analysis showed association to immune processes and metabolism. For BG, only 21 DEG were observed at d31, these DEGs were associated to signal transduction and sympathetic nerve pathway. For GOS, 816 DEG were observed for GOS at d1, and 77 at d31, where DEGs at d1 were associated to immune processes. For the neonatal administration route, MCFA showed 94 DEG and GOS 6 DEG. Where DEGs in MCFA were mainly associated to cell adhesion processes. When comparing the administration routes directly between treatment groups, we observed significant differences in alpha diversity parameters, observed species at d31 for MCFA, Shannon for GOS, as well as for beta diversity in GOS. For MCFA 515 DEG were observed, for BG 503 DEG, and for GOS 996 DEG. Where for MCFA most pathways were associated to immunological processes, BG showed more metabolism, and GOS mainly metabolism with a few immunological processes. The type of intervention and the administration route influence gut functionality of the piglets. MCFA administration led to a more differentially orchestrated response when comparing the neonatal and maternal administration route then the other two additives. This implies that for each nutritional intervention in early life of a pig the optimal route of administration needs to be determined.
... The effects of organic acids on intestinal morphology are similarly variable. Some studies report no changes in gut structure with organic acid supplementation [55,56], while others demonstrate improvements in villous height-to-crypt depth ratios, indicating enhanced nutrient absorption [57,58]. ...
Article
Full-text available
Weaning in piglets presents significant physiological and immunological challenges, including gut dysbiosis and increased susceptibility to post-weaning diarrhoea (PWD). Abrupt dietary, environmental, and social changes during this period disrupt the intestinal barrier and microbiota, often necessitating antimicrobial use. Sustainable dietary strategies are critical to addressing these issues while reducing reliance on antimicrobials. Reducing dietary crude protein mitigates the availability of undigested proteins for pathogenic bacteria, lowering harmful by-products like ammonia and branched-chain fatty acids, which exacerbate dysbiosis. Organic acid supplementation improves gastric acidification, nutrient absorption, and microbial balance, while also serving as an energy-efficient alternative to traditional grain preservation methods. Increasing intestinal butyrate, a key short-chain fatty acid with anti-inflammatory and gut-protective properties, is particularly promising. Butyrate strengthens intestinal barrier integrity by upregulating tight junction proteins, reduces inflammation by modulating cytokine responses, and promotes anaerobic microbial stability. Exogenous butyrate supplementation via salts provides immediate benefits, while endogenous stimulation through prebiotics (e.g., resistant starch) and probiotics promotes sustained butyrate production. These interventions selectively enhance butyrate-producing bacteria such as Roseburia and Faecalibacterium prausnitzii, further stabilising the gut microbiota. Integrating these strategies can enhance gut integrity, microbial resilience, and immune responses in weaned piglets. Their combination offers a sustainable, antimicrobial-free approach to improving health and productivity in modern pig production systems.
... Consequently, exploring non-antibiotic alternatives to effectively mitigate weaning stress and support healthy growth has become a crucial focus in animal science research. Dietary supplementation with amino acids, fatty acids, and other nutrients has been shown to benefit weanling piglets [6,7]. The nonprotein amino acid γ-aminobutyric acid (GABA) has gained attention for its diverse roles in regulating intestinal motility, carcinogenesis, pain management, and inflammation [8,9]. ...
Article
Full-text available
The nonprotein amino acid γ-aminobutyric acid (GABA) can enhance intestinal function in piglets; however, the mechanisms involved are not yet fully understood. To explore the effects of GABA and its underlying mechanisms, weanling piglets were randomly assigned to three groups, receiving either a basal diet or a basal diet supplemented with GABA (80 mg/kg or 120 mg/kg). The results demonstrated that dietary GABA improved growth performance and reduced diarrhea incidence (p < 0.05). Additionally, GABA supplementation decreased the serum and intestinal levels of pro-inflammatory cytokines (p < 0.05), and improved intestinal morphology. Multi-omics analyses were employed to explore the alterations caused by GABA supplementation and elucidate the related mechanisms. Microbiota profiling revealed improved beta-diversity and changes in the composition of ileal bacteria and fungi. Amino acid metabolism, lipid metabolism, and digestive processes were primarily enriched in the GABA group according to metabolomics analysis. A transcriptome analysis showed significant enrichment in ion transmembrane transport and nutrition absorption and digestion pathways in the ileum. Furthermore, increased lipase and trypsin activity, along with the elevated expression of tight junction proteins confirmed the beneficial effects of GABA on intestinal nutrient metabolism and barrier function. In conclusion, dietary 80 mg/kg GABA supplementation improved nutrient digestion and absorption and intestinal function in weanling piglets.
... These substances can offer instant energy and may also provide physiological advantages that extend beyond their calorie content since they affect the composition of the intestinal microbiota and have inhibitory effects on intestinal bacterial concentrations (Zentek et al., 2011). In addition, studies have shown support of MCFAs on the integrity of the intestine, increasing the villi length, reducing the crypt depth in the small intestine, and decreasing intestinal inflammation (Ferrara, Tedin, Pieper, Meyer, & Zentek, 2017). ...
... The effect of using a mixture of acids (as the blend used in this study) is generally better than that of a single acid due to synergism and their dissociation properties at different locations within the digestive tract (Huang et al., 2015). Fumaric acid, lactic acid, capric acid and caprylic acids didn't prove to have an effect on the morphometry of jejunum in piglets (Ferrara et al., 2017). In the same study an increasing number of potential effector cells (principally CD3+ cytotoxic T lymphocytes) was found. ...
Article
Full-text available
Weaning is a stressful period in a piglet’s life, when many morphological and functional changes occur in the gastrointestinal tract. Examples are an alteration of the barrier function and an increase in production of cytokines, suggesting the immune system of the GIT is activated after weaning. The goal of this study was to evaluate the effect of a commercially available feed additive based on short and medium chain fatty acids on supporting intestinal health and reduce inflammation in the intestine of post-weaning piglets. Seventy piglets were divided over 2 treatments, a control diet and a blend of short and medium chain fatty acids, including butyrate. On day 0, 15, 30 and 45 after weaning blood samples and intestinal tissue samples were collected from sacrificed piglets. Blood and tissue samples were analysed for cytokine gene expression, and tissue samples from jejunum and ileum were analysed on morphology and gene expression of tight junctions. Results show that the use of a blend of short and medium chain fatty acids, including butyrates, improved intestinal morphology, measured by villous length and v/c ratio, reduced expression of tight junction proteins and influenced cytokine production in the intestine, implicating that the use of organic acids as feed additives potentially can reduce damage caused by weaning to the intestinal barrier and due to the higher absorption capacity of the villi potentially improve nutrient absorption.
... However, the roles of IELs in porcine immune function are poorly understood. Weaning piglet may exhibit a significantly increased total number of IELs (Ferrara et al., 2017), and this increase in IEL count may be due to the increased diversity of gut microbiota in piglets (Zheng et al., 2020). Unfortunately, studies on porcine MAIT cells are limited. ...
Article
Full-text available
With gradual ban on the use of antibiotics, the deficiency and excessive use of trace elements in intestinal health is gaining attention. In mammals, trace elements are essential for the development of the immune system, specifically T-cell proliferation, and differentiation. However, there remain significant gaps in our understanding of the effects of certain trace elements on T-cell immune phenotypes and functions in pigs. In this review, we summarize the specificity, development, subpopulations, and responses to pathogens of porcine T cells and the effects of functional trace elements (e.g., iron, copper, zinc, and selenium) on intestinal T-cell immunity during early-life health in pigs. Furthermore, we discuss the current trends of research on the crosstalk mechanisms between trace elements and T-cell immunity. The present review expands our knowledge of the association between trace elements and T-cell immunity and provides an opportunity to utilize the metabolism of trace elements as a target to treat various diseases.
Article
Full-text available
This study aimed to evaluate the dietary administration of a blend composed of carvacrol, tannic acid derived from Castanea sativa mill and Glycyrrhiza glabra, medium chain fatty acids (MCFAs) glycerides for weanling piglets. An in vitro digestion followed by total phenolic content (TPC) and total antioxidant activity (TAC) assessment was performed before the in vivo application. At weaning, a total of 210 piglets were randomly allocated to two experimental treatments (7 replicates/15 piglets for each replicate). Control group (CTR) was fed a standard basal diet while the treated group (T) was fed the basal diet mixed with 1.500 mg/kg of blend. After in vitro digestion, TPC and TAC evidenced peaks at the end of oral and gastric phases in comparison to the intestinal one in line with the high content of phenolic compound (P < 0.05). Treatment conditioned body weight and average daily gain (P < 0.05), fecal score on 6, 7, and 8 d after weaning (P < 0.05). At 35d, the T group showed a decrease in salivary cortisol compared to CTR (P < 0.05). Duodenum and jejunum sections of T piglets revealed higher villi (P < 0.05), deeper crypts (P < 0.01), and increased V/C ratio (P < 0.01). CTR showed a higher expression of duodenal Occludin (P < 0.05). Jejunal E-cadherin and Occludin were more expressed in T jejunum sections (P < 0.05). Twelve differentially abundant genera were identified in T group caecal samples. Potentially harmful Clostridium sensu stricto 13 was reduced by the treatment (P < 0.05). In conclusion, the tested blend positively affected salivary stress markers and the gut health of weaned piglets.
Article
Full-text available
This study was aimed at determining the effect of Vernonia amygdalina leaf meal (VALM) extract in drinking water on the faecal microbial load on post-weaning diarrhea occurrence in piglets using different agar. Forty-five piglets of Large white X Duroc breed were given Vernonia amygdalina (VA) through water infusion. Three levels of 0.0g, 1.2g, and 2.4g of VA per 1000ml of clean drinking water designated as T1, T2, and T3 were used. Nine piglets weaned at different ages of 2nd, 3rd, 4th, 5th, and 6th week of age were randomly allotted to three treatments with each treatment replicated three times with a piglet per replicate in a completely randomized design. The different agar used are Macconkey Agar, Eosin Methylene Blue (EMB) Agar, and Nutrient Agar. These agars were used for 4days each to determine the faecal microbial load. The experiment lasted 28 days. Results showed that the bitter leaf contain high Alkaloids, medium Tannins, Saponins Flavonoids, and low amounts of Terpenoids and Phenols while Cardiac Glycosides were not determined. Bitter leaf showed the power of antibacterial activity for the reduction of faecal microbial load. Bitter leaf as a feed additive in the drinking water of piglets caused a significant (P<0.05) reduction in the faecal microbial load for piglets given 1.2g and 2.4g of VA per 1000ml of drinking water. It can be concluded that bitter leaf extract in drinking water can be used as phyto-additive for piglets to reduce faecal microbial load.
Preprint
Full-text available
Post Weaning Diarrhea (PWD) is the most important multifactorial gastroenteric disease of the weaning in pig livestock. Phytogenic (PHY) natural extracts are largely studied as alternatives to antibiotic treatments in combating the global concern of the antimicrobial resistance. The aim of this study was to evaluate the protective effect of innovative phytogenic premix with or without short and medium chain fatty acids (SCFA and MCFA) in O138 E. coli challenged piglets. Twenty-seven weaned piglets were allotted into four groups receiving different diets according to the following experimental doses: PHY1 (n=7) fed basal diet supplemented with 0.2% of phytogenic premix, PHY2 (n=7) fed basal diet supplemented with 0.2% of phytogenic premix added with 2000 ppm of SCFA and MCFA. After 6 days of feeding experimental diets, animals were challenged (day 0) with 2x10 ⁹ CFU/dose of E. coli and CTRL group fed basal diet was divided at day 0 into positive (CTRL+; n=6) and negative control group (CTRL-; n=7). Body weights were recorded at -14, -6, 0, 4 and 7 days and the feed intake was recorded daily. E. coli shedding was monitored for 4 days post-challenge by plate counting. Fecal consistency was registered daily by a four-point scale (0-3; diarrhea >1) during the post-challenge period. Tissue samples were obtained for gene expression and histological evaluations at day 7 from four animals per group. Lower average feed intake was observed in CTRL+ compared to PHY2 and CTRL during the post-challenge period. Infected groups showed higher E. coli shedding compared to CTRL- during the 4 days post-challenge ( p <0.01). PHY2 showed lower frequency of diarrhea compared to PHY1 and CTRL+ from 5 to 7 days post-challenge. No significant alterations among groups were observed in histopathological evaluation. Duodenum expression of occludin tended to be lower in challenged groups compared to CTRL- at 7 days post-challenge ( p =0.066). In conclusion, dietary supplementation of PHY plus SCFA and MCFA revealed encouraging results for diarrhea prevention and growth performance in weaned piglets.
Article
Full-text available
Pigs experience biological stress such as physiological, environmental, and social challenges when weaned from the sow. The process of weaning is one of the most stressful events in the pig's life that can contribute to intestinal and immune system dysfunctions that result in reduced pig health, growth, and feed intake, particularly during the first week after weaning. Technological improvements in housing, nutrition, health, and management have been used to minimize some of the adverse effects of weaning stress, but a greater understanding of the biological impact of stress is needed to improve strategies to overcome weaning stress. The focus of this review paper is to briefly describe how the biological stress associated with weaning impacts intestinal morphology, structure, physiology, and intestinal immune responses that can impact subsequent production efficiencies such as growth, intake, morbidity, and mortality.
Article
Full-text available
Porcine γδ T cells have two levels of TCRγδ expression. Whereas TCRγδ(med) cells are mostly CD2(+)CD8(-) and CD2(+)CD8(+), TCRγδ(hi) cells are highly enriched for CD2(-)CD8(-). This distribution is independent of bacterial colonization and it is already established in the thymus prior to export of γδ cells to the periphery. Sorting and cultivation experiments revealed that CD2(-)CD8(-) γδ cells are unable to acquire CD2 and CD8, whereas CD2(+) subsets can gain or loose CD8. There is also differential susceptibility for proliferation between CD2(+) and CD2(-) γδ cells. Although CD2(-)CD8(-) almost do not proliferate, proliferation of CD2(+)CD8(-) and CD2(+)CD8(+) is substantial. Population of CD2(-) γδ cells is also absent in CD1(+) immature thymocytes. Additionally, subpopulations of CD2(+) and CD2(-) γδ cells in the thymus differ in expression of auxiliary surface molecules such as CD25, CD45RA/RC, and MHC class II. Moreover, TCRγδ(hi) cells can generate TCRγδ(med) cells but never the opposite. The only exception is the thymus, where a few TCRγδ(med) cells can be induced to TCRγδ(hi) but only under IL-2 influence. The repertoire of TCRδ is polyclonal in all subsets, indicating that there is the same extent of diversification and equal capability of immune responses. Results collectively indicate that CD2 expression determines two lineages of γδ cells that differ in many aspects. Because CD2(-) γδ cells are missing in the blood of humans and mice but are obvious in other members of γδ-high species such as ruminants and birds, our findings support the idea that circulating CD2(-) γδ T cells are a specific lineage.
Conference Paper
Our recent Studies of structure and function of gastrointestinal tract mucosa revealed that the domestification of Sus scrofa corresponds with the significant slowing of the organ development. On top of genetic potential, the nutritional factors (or more precisely - lack of certain biologically active substances in the feed of pregnant sows) arc responsible. Moreover, feeding neonates with milk replacers instead of mother's milk further slows down the development. This is manifested by reduced mitotic activity in the crypts and enhanced apoptosis of enterocytes. The negative effects consist of slower replacement of fetal type, vacuolated enterocytes to adult type enterocytes, modified profile of brush border enzymes, alterations in intestinal mucosa barrier, higher susceptibility to infectious agents, and many others. On the other hand, farmers in order to intensify the production, shorten the suckling period imposing the neonatal piglets to be weaned at 3-4 weeks of life and even earlier. Altogether, it makes the weaning disorders one of the most important problems in pig husbandry, and the mortality of piglets in the leading pig-producing countries still reaches 10%. A number of strategies have been developed to counteract the post-weaning problems. One of them is to stimulate the development of the gastrointestinal tract of the neonate by supplementation of the sow diet with certain biologically active substances and plants. The other idea is to speed up the postnatal development of the gut mucosa for example by plant lectins. Lessons from pig studies can be also useful in human nutrition and medicine since the development of porcine gastrointestinal tract shows a great similarity to that of humans.
Article
Two experiments were conducted to evaluate effects of dietary solid mixture of organic acids and medium chain fatty acids (MCFA) as a replacement of zinc oxide (ZnO) on growth, digestibility and immunity of weanling pigs. In Exp. 1, the diet with supplemental antibiotics (containing pure Colistin sulfate and Enramycin, respectively, at 0.02. g/kg diet) and ZnO (2.5. g/kg diet) was used as the control (CON) group, and another diet without supplemental ZnO was prepared by inclusion of the SF3 (containing calcium formate, calcium lactate, citric acid and MCFA at 340, 160, 70 and 130. g/kg, respectively) at 3. g/kg diet. A total of 120 crossbred piglets (21-day old, 6.36 ± 0.80. kg) were fed the same commercial prestart diet for one week and then fed the CON or SF3 diet for four weeks (ZnO was no longer supplemented in the CON diet during the second two-weeks), with 6 replicate pens (10 piglets/pen) per diet. Compared with the CON-fed piglets, the SF3-fed piglets had higher (P< 0.001) feed intake during the first two experimental weeks, higher (P< 0.05) body weight at the end of each of the four experimental weeks, and lower (P< 0.01) ratio of feed to gain during the first experimental week. In Exp. 2, diets were the same as in Exp. 1, except that chromium oxide was included at 3. g/kg diet at the expense of corn to evaluate apparent ileal digestibility (AID) of dietary amino acids (AA). Fourteen crossbred piglets (28-day old, 9.05 ± 1.38. kg) were housed individually in metabolism cages, fed the CON diet for one week and then fed the CON or SF3 diet for two weeks with 7 pigs (4 female and 3 male) per diet. Compared with the CON-fed piglets, the SF3-fed piglets had higher (P< 0.05) AID of a majority of AA, lower (P< 0.05) plasma tumor necrosis factor alpha (TNF-α) but higher (P< 0.05) plasma immunoglobulin-G concentrations, more (P< 0.05) ileal and rectal Lactobacillus content, and upregulated (P< 0.05) jejunal AA transporters (EAAT3 and CAT2) and regulatory cytokine (transforming growth factor beta) expression but downregulated (P<. 0.05) jejunal proinflammatory cytokine (TNF-α) expression. Taken together, dietary supplementation with a mixture of organic acids and MCFA contributed more than ZnO to growth of weanling pigs under commercial conditions. The improved growth of pigs was associated with increased AA digestibility and immunity. The stimulated regulatory cytokine expression and downregulated proinflammatory cytokine expression might be associated with increased proliferation of Lactobacillus following organic acids consumption.
Article
Organic short and medium chain fatty acids are used in diets for piglets because they have an impact on the digestive processes and the intestinal microbiota. In this study, 48 pens (2 piglets/pen) were assigned randomly to 4 diets, without additive (control), with organic acids (OA; 0.416% fumaric and 0.328% lactic acid), with medium chain fatty acids (MCFA; 0.15% caprylic and capric acid), and a combination of OA and MCFA, to assess changes in the gastrointestinal microbiota with 12 pens per diet. Eight to nine piglets from each group were euthanized after 4 wk. Organic acids, MCFA, and pH in the digesta were determined and the intestinal microbiota was quantified by real-time PCR. The different diets had no effect on the growth performance. Concentration of added fumaric acid was below the detection limit in the upper small intestine whereas the concentration of lactic acid in the digesta was not affected by the treatments. The added MCFA was recovered in the MCFA treated groups in the stomach, but the concentrations declined in the upper small intestine. Concentration of short chain fatty acids was reduced in the colon digesta in piglets fed diets with OA compared with those fed unsupplemented diets (P = 0.029). The MCFA resulted in a pH reduction of the digesta, likely because of the effect on bacterial acid production. The addition of OA increased cell counts of Bacteroides-Porphyromonas-Prevotella group and clostridial clusters XIVa, I, and IV in the stomach, the clostridial cluster XIVa in the jejunum, and Bacteroides-Porphyromonas-Prevotella in the ileum and reduced counts of Streptococcus spp. in the colon (P < 0.05). The MCFA induced only minor changes in the gastrointestinal microbiota but increased cell counts for the Escherichia-Hafnia-Shigella group in the jejunum and the clostridial cluster XIVa in the colon digesta (P < 0.05). In the colon of piglets fed diets with organic OA, reduced mean cell counts of STb (est-II) positive Escherichia coli were found. In conclusion, OA and MCFA had effects on the intestinal microecology in piglets. The decrease of the intestinal pH and the reduction of E. coli virulence genes by OA could make the combination of short chain fatty acids and MCFA as interesting gut flora modifiers, which can eventually prevent postweaning diarrhea.
Article
This study aimed at studying the effect of age and Enterococcus faecium NCIMB 10415 on jejunal physiological measures in suckling and weaned piglets. A total of 112 piglets from 16 landrace sows were used. Sows received either no probiotic (n = 8, control) or were fed a diet with 4.2 to 4.3*106 cfu/g E. faecium NCIMB 10415 (n = 8, EF) from 28 days ante partum onwards. Piglets were offered creep feed beginning at 12 d of age and piglets were weaned to a pre-starter diet at 26 d of age. For EF piglets, the creep feed (pre-starter) and starter diets contained 5.1×106 cfu/g and 3.6×106 cfu EF/g, respectively. Twenty-seven randomly selected piglets from each group were euthanized at 12 (n = 6), 26 (n = 6), 34 (n = 7), and 54 d (n = 8) of age and a 45 cm mid-jejunal segment was taken. Histomorphometry was performed and relative transcript abundance of caspase-3, proliferating cell nuclear antigen, intestinal alkaline phosphatase (IAP), lactase-phlorizin hydrolase (LPH), sucrase (SUC), aminopeptidase-N (APN) and sodium/glucose cotransporter 1 (SGLT1) was determined. Lactase (LAC), maltase (MAL), SUC, APN and IAP activities were measured in brush border membranes. The EF treatment did not affect (P > 0.05) performance variables like the pre- or post-weaning daily weight gain, feed intake, incidence of diarrhoea and ileal and total tract nutrient digestibility, except the feed conversion ratio which was reduced (P = 0.033). Jejunal villus length and crypt depth did not differ between groups (P > 0.05) but increased age-dependently (P = 0.003 and P < 0.001, respectively). Feeding EF did not markedly influence brush border enzyme expression or activity. Enzyme activity increased (SUC, MAL) or decreased (LAC, IAP) with age reflecting maturation and adaptation to creep feed (P < 0.001). The gene expression of SUC, SGLT1 and IAP changed over time (P = 0.012, P = 0.003 and P = 0.013). Furthermore, gene expression was not affected by treatment. The current study suggests that EF did not markedly affect physiological variables associated with digestive function. Age of piglets seems to be the main factor determining function and morphology of the jejunal tissue, reflecting both maturation and adaptation to creep feed.
Article
In a first set of experiments, it was examined if, with four selected medium-chain fatty acids (MCFAs) (C6:0–C12:0) containing fat sources (coconut, MCTAG1, MCTAG2, butter) and six lipases, under gastric simulated conditions (pH 3 to 6, 3 h incubation, 37°C, with complex substrate), appropriate amounts of MCFAs could be liberated, which could provide a valuable alternative for nutritional antibiotics used in piglet nutrition. Depending on the conditions applied, up to 20% of the MCFAs could be enzymatically released into the medium. In a second set of experiments, the generation and antimicrobial effects of MCFAs from three selected MCFA-containing fat sources (coconut, MCTAG1 and MCTAG2) and one appropriate microbial lipase, applied under different combinations and concentrations, were studied. A minimal concentration of 0.35 g per 100 g incubation fluid or 0.025 M MCFAs in the medium (e.g., stomach, proximal gut) appears to be necessary to obtain a significant (>10-fold) suppression of the flora. This amount can be obtained by choosing an appropriate combination of sources and doses of fat and lipases. The in vitro protocol used offers an excellent tool for the screening of the numerous combinations of MCFA-containing fat sources and lipolytic enzymes for their usefulness as feed supplements which comply with the above conditions, which are generally accepted as the prerequisite for obtaining growth promotion.
Article
In a first set of experiments, it was examined if, with four selected medium-chain fatty acids (MCFAs) (C6:0–C12:0) containing fat sources (coconut, MCTAG1, MCTAG2, butter) and six lipases, under gastric simulated conditions (pH 3 to 6, 3 h incubation, 37°C, with complex substrate), appropriate amounts of MCFAs could be liberated, which could provide a valuable alternative for nutritional antibiotics used in piglet nutrition. Depending on the conditions applied, up to 20% of the MCFAs could be enzymatically released into the medium. In a second set of experiments, the generation and antimicrobial effects of MCFAs from three selected MCFA-containing fat sources (coconut, MCTAG1 and MCTAG2) and one appropriate microbial lipase, applied under different combinations and concentrations, were studied. A minimal concentration of 0.35 g per 100 g incubation fluid or 0.025 M MCFAs in the medium (e.g., stomach, proximal gut) appears to be necessary to obtain a significant (>10-fold) suppression of the flora. This amount can be obtained by choosing an appropriate combination of sources and doses of fat and lipases. The in vitro protocol used offers an excellent tool for the screening of the numerous combinations of MCFA-containing fat sources and lipolytic enzymes for their usefulness as feed supplements which comply with the above conditions, which are generally accepted as the prerequisite for obtaining growth promotion.