Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/331446449

Arabinoxylan structural characteristics, interaction with gut microbiota and


potential health functions

Article  in  Journal of Functional Foods · March 2019


DOI: 10.1016/j.jff.2019.02.007

CITATIONS READS

0 118

6 authors, including:

Hong Chen
Sichuan Agricultural University
61 PUBLICATIONS   483 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

human health View project

All content following this page was uploaded by Hong Chen on 11 March 2019.

The user has requested enhancement of the downloaded file.


Journal of Functional Foods 54 (2019) 536–551

Contents lists available at ScienceDirect

Journal of Functional Foods


journal homepage: www.elsevier.com/locate/jff

Arabinoxylan structural characteristics, interaction with gut microbiota and T


potential health functions

Zhuoyun Chena,1, Shanshan Lia,1, Yuanfang Fua, Cheng Lia, Daiwen Chenb, Hong Chena,
a
College of Food Science, Sichuan Agricultural University, Ya’an, Sichuan 625014, China
b
Institute of Animal Nutrition, Sichuan Agricultural University, Postcode 611130, Chengdu, Sichuan, China

A R T I C LE I N FO A B S T R A C T

Keywords: Arabinoxylans are the main type of noncellulose polysaccharide in cereals and grasses. They are also important
Dietary fiber functional ingredients in baked products. Arabinoxylans derived from different sources or extracted in different
Physiological functions ways may exhibit different structures which give rise to various physiological functions and exert diverse im-
Structural features pacts on gut microbiota. This review summarizes the most current researches on arabinoxylans’ beneficial effects
Fermentation
to human health, their prebiotic effects, the influences of structural features on short-chain fatty acids pro-
Prebiotic effects
Antioxidant effect
duction as well as on physiological functions. We aim to identify arabinoxylans’ primary structural features that
related to each functional property and also provide the explaination to the inconsistent antioxidant capacity of
arabinoxylans with similar structural features.

1. Introduction techniques (Fadel et al., 2017; Fry, 2004; Izydorczyk & Biliaderis, 1995;
Yadav, Kale, Hicks, & Hanah, 2017). The structures of AXs, however,
Arabinoxylans (AXs) are the main kind of hemicelluloses in many originated from different raw materials or extracted by different
cereal grains. They are mostly originated from the bran and starchy methods are not highly consistent. Each may has different unique
endosperm, and can’t be degraded by mammalian enzymes (Fadel et al., structural features (different sugar composition, molecular weight
2017; Zhou et al., 2010). AXs demonstrate many functional properties (MW), type of branching, binding, etc.). For instance, compared with
in food production and physiological processes during metabolism. For AXs from barley, AXs from banana peel hemicellulose or from corn hull
instance, they can strengthen the immune system (Biliaderis & contain higher galactose or glucuronic acid while enzymatic treatment
Izydorczyk, 2007; Glitso, Jensen, & Bach Knudsen, 2000; Izydorczyk & produced AXs have lower molecule weight and with higher ferulic acid
Dexter, 2008; Qiang, Chao, & Wan, 2009), lower the possibility of the (FA) content than alkali treated ones (Ogawa, Takeuchi, & Nakamura,
occurrence of diseases (e.g. cardiovascular diseases, diabetes type 2, 2005; Samuelsen, Rieder, Grimmer, Michaelsen, & Knutsen, 2011; Zhou
obesity and colon cancer), decrease the organ damage especially for et al., 2010).
liver as well as promote the number of probiotic bacteria growing in gut Lots of studies have shown that AXs with different structural fea-
(Biliaderis & Izydorczyk, 2007; Chen et al., 2018). It is believed that the tures exhibit different biological activities. There is a strong correlation
change of intestinal microbiota stimulated by AXs improves minerals between molecule structure and physiological functions for AXs (Ayala-
absorption and resists the invasion of pathogen in the gut (Broekaert Soto, Serna-Saldívar, Welti-Chanes, & Gutierrez-Uribe, 2015; Gemen,
et al., 2011). de Vries, & Slavin, 2011; Geraylou et al., 2013; Malunga, Izydorczyk, &
AXs can be derived from a wide range of sources including cereals Beta, 2017; Mendis, Leclerc, & Simsek, 2016; Sarma et al., 2018; Zhang,
such as maize, rye, barley, oats, sorghum, wheat, rice, and other plants Li, Smith, & Musa, 2015). The aim of this article is to summarize the
such as banana. Likewise, varieties of extraction methods have been health benefits of AXs occurred in the physiological processes. Because
studied to produce AXs, for example, water, alkaline, enzyme extrac- some of the health benefits are related to gut microbiota, we also sum
tions, and physical treatments and various combination of the up the interaction between AXs and gut microbiota. In the meantime,

Abbreviations: AXs, arabinoxylans; MW, molecular weight; FA, ferulic acid; DP, degree of polymerization; DS, degree of substitution; AXOS, arabinox-
ylooligosaccharides; SCFAs, short chain fatty acids; PUL, polysaccharide utilization loci; Sus, starch utilization system; LDL, low-density lipoprotein; BA, bile acids;
GLP-1, glucagon-like peptide 1; GLP-2, glucagon-like peptide 2; Igs, immunoglobulins; WEAX, water extractable AXs

Corresponding author.
E-mail address: chenhong945@sicau.edu.cn (H. Chen).
1
Zhuoyun Chen and Shanshan Li contributed equally to this article.

https://doi.org/10.1016/j.jff.2019.02.007
Received 27 October 2018; Received in revised form 4 February 2019; Accepted 4 February 2019
1756-4646/ © 2019 Elsevier Ltd. All rights reserved.
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

Xyl - Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl –Xyl – Xyl – Xyl
2 3 3 2 3 2

1 1 1
β-L-Ara 1 3 α-L-Ara 1 3α-L-Ara Ara 1 Ara 1
5 Glca 5 Ara
O
O

Gal Glu Ara


1 1 1

2 2 3
Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl –Xyl – Xyl – Xyl – Xyl
2 2 2 2

1 1 1
Gal – Ara Ara – Ara
Xyl – Ara

Fig. 1. The schematic diagram of AX main structural characteristics. The structure of AX can vary and are related to the raw materials or extraction methods (Zhang,
Li, et al., 2015).

this article focuses on the structural differences of AXs which influence different types of molecular structures and spatial arrangements of AXs
the health benefits. Several current articles have demonstrated that AXs (Döring, Jekle, & Becker, 2016). In addition, it has been shown that
lacking of structural differences, however, resulted in different anti- small contents of other sugar residues such as glucuronic acids, 4-O-
oxidant effects, the possible mechanisms are assumed in this review. methyl glucuronic acid, or short oligomers consisting of L-arabinose, D-
xylose, D-galactose, D-glucose, and/or uronic acids can substitute hy-
2. The molecular structure of AXs drogen atom at O-2 position of xylose residues, while acetyl groups
substitute at O-2 and/or O-3 of xylose residues in AXs (Courtin, 2011).
The main structural characteristics of AXs are summarized in Fig. 1. Hydroxycinnamic acid derivatives, such as ferulic, coumaric, sinapic,
The linear main chain of AXs is constituted of a β-1.4-D-xylopyranosyl dehydrodiferulic acids, linked to arabinofuranosyl residues through
backbone with L-arabinofuranosyl substituted at position 2 and/or 3 of ester linkages with the 5-OH group (Courtin, 2011; Izydorczyk &
the furan cycle. Consequently, there are four types of substitution Dexter, 2008), can shape the dehydrodimer and dehydrotrimer isomers
pattern of xylopyranosyl: unsubstituted, monosubstituted at O-2, to potentially crosslink polysaccharide chains and polysaccharide-pro-
monosubstituted at O-3, or disubstituted at O-2.3 (Biliaderis & tein (Dervilly-Pinel, Tran, & Saulnier, 2004; Dervillypinel, Rimsten,
Izydorczyk, 2007; Izydorczyk & Biliaderis, 1992; Vanhamel, Cleemput, Saulnier, Andersson, & Aman, 2001; Izydorczyk & Macgregor, 2000;
Delcour, Nys, & Darius, 1993). Mccleary & Prosky, 2008).
Xylopyranosyl with different substitution patterns has a diverse The molecular structure of AXs is also related to the extraction
ratio of arabinose to xylose, which plays an important role in depicting method applied. AXs can be extracted using chemical, enzymatic, or
the molecular structure of AXs. For example, it affects the hydrogen physical treatments. Several types of research showed that alkaline
bonds leading to the change of spatial arrangement of AXs (Courtin & treatment affected the functional group of AXs, lowered FA content,
Delcour, 2002; Köhnke, Ostlund, & Brelid, 2011). The arabinofuranosyl and maintained the native molecule weight of AXs. On the other hand,
molecules of AXs presented as monosubstituted residues are mostly and enzymatic treatment produced lower molecule weight AXs but with
mainly monosubstituted at O-3, except isolated barley and rye fractions higher FA content (Zhou et al., 2010). While physical treatments such
with a large amount of arabinofuranosyl molecules monosubstituted at as extrusion cooking reduced the molecule weight of AXs because it
O-2 (Cja, Delcour, Verbruggen, & Gruppen, 1995; Ebringerová, lowered the degree of polymerization (DP) by breaking AXs glycosidic
Hromádková, Petráková, & Hricovíni, 1990; Izydorczyk & Biliaderis, bonds (Nyman, 2002).
1993). As for the conformation of L-arabinofuranosyl, it has been re- The structure of AXs depends also on the raw material that they are
ported that the xylan backbone of new AXs is substituted by a mixture originated from. AXs derived from banana peel hemicellulose and from
of α- and β-anomers arabinose residues (Das, Maiti, Maiti, & Islam, corn hull contained galactose or glucuronic acid (Ogawa et al., 2005;
2013) although most kinds of xylan backbone were only substituted by Zhang et al., 2004). AXs derived from the green leaves of Litsea glutinosa
α-L-arabinofuranosyl. Additionally, these oligomeric side chains are had a special type of branching (Das et al., 2013). Additionally, Kodo
attached to each other via 1 → 2, 1 → 3, and 1 → 5 linkages, resulting in millet contained comparatively low branched AXs with a high amount

537
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

of FA, p-coumaric, and caffeic acid, while glucoarabinoxylans extracted however, is still unclear. The Bacteroidetes downstream metabolism was
from high tannin sorghum brans contained a high content of total fermented by Firmicutes and Actinobacteria, which played important
phenolic especially tannin. Altogether, it can be concluded that raw roles in the further degradation by gram-positive PULs, analogous to
materials and extraction method determine the molecule weight, sugar the PUL systems in Bacteroidetes (Sheridan et al., 2016). Gram-positive
composition, degree of substitution (DS), type of branching and total PULs encoded a range of transporters, regulatory proteins, and Carbo-
phenolic content of AXs. hydrate-Active Enzymes to bind, degrade and transport AXs with low
DP and downstream metabolisms. The main chain of AXOS was hy-
3. AXs structure related to physicochemical properties drolyzed first, and then α-1,3- linked arabinofuranosyl residues from
AXOS were removed by glycoside hydrolases to produced xylose and
Compared with insoluble AXs, soluble AXs have much more func- arabinose (Michlmayr et al., 2013). L. acidophilus, L. brevis, and Bifi-
tional properties. For instance, the existence of soluble AXs in bread dobacterium species, for instance, produced enzymes to perform these
leads to improvement of dough parameters as well as bread quality processes (Michlmayr et al., 2013; Rivière et al., 2014). The produced
(Buksa, Nowotna, Praznik, & Gambuś, 2012). At the same time, soluble xylose then was used by some species of Bifidobacterium, such as Bi.
AXs increase water content and lower the retention times of digesta in catenulatum and Bi longum. Arabinose was used by Bi. Dentium to gen-
pig thereby promoting regularity and digestive health (Zhang, erate SCFAs. Likewise, Roseburia sp. also has been shown to be able to
Williams, et al., 2015). It has been reported that the solubility of AXs is ferment xylose to SCFAs (Ndeh & Gilbert, 2018). AXs stimulate the
determined by its structure. Arabinose: xylose ratio and the present of growth of gut microbiota species which affect the composition of
FA are associated with the solubility. The unsubstituted parts of AXs SCFAs. Acetate is the dominant SCFAs in AXOS treatment. After the
tend to aggregate through hydrogen bonds, leading to a lower solubi- experiment in fish, (Geraylou et al., 2013) found that the abundant of
lity. An arabinose: xylose ratio lower than about 0.43 causes a drastic acetate producers, such as Clostridium spp. and lactic acid bacteria
drop in solubility indicating that higher arabinose: xylose ratio exerted elevated after AXOS treatment, which could be one of the reasons of the
a positive effect on solubility (Andrewartha, Phillips, & Stone, 1979; higher amount of acetate. It has been shown that AXOS is utilized by
Courtin & Delcour, 2002; Köhnke et al., 2011; Mccleary & Prosky, cross-feeding bifidobacteria and butyrate-producing colon bacteria, such
2008). The bound FA exhibits the cross-linking effect in each AX, and as Faecalibacterium prausnitzii (clostridialclusterIV) and Anaerostipes,
consequently, decreases the solubility. Eubacterium, and Roseburia species (clostridialclusterXIVa), to perform
Dhital, Dolan, Stokes, and Gidley (2014) investigated that AXs in- the butyrogenic effects (Audrey, Marija, David, Frédéric, & Luc, 2016).
crease solution viscosity in vitro, thus, soluble AXs were likely thought At the same time, since long-chain AXs supplementation was not ab-
to form viscous digesta in the stomach as well as intestinal. The visc- sorbed in small intestinal and would progress to the caecal, it changed
osity of AXs can change lipid and glucose metabolism. Some of the the predominant habitat of the mucin-degrading Akkermansia mucini-
proposed mechanisms include delaying gastric emptying, slowing the phila from the caecum to more distal regions, where Akkermansia mu-
absorption and impacting the synthesis of enzyme or hormone (Fadel ciniphila could convert mucins to propionate in humanized rats (Van
et al., 2017; Hartvigsen et al., 2014). The molecular structure of AXs den Abbeele et al., 2011). Besides, the high amount of SCFAs inhibited
affects the viscosity properties to some extent. Comparing AXs from the growth of Bacteroides species (Broekaert et al., 2011).
wheat bran to that from two rye breads, the AXs from bran had a higher
degree of branching, which led to higher viscosity, indicating that the 4.2. AXs structure and microbita fermentation
degree of branching positively related to viscosity (Christensen et al.,
2013). Similarly, AXs with higher MW have higher viscosity Researches show that the structure of AXs is related to the gut mi-
(Christensen et al., 2013). Likewise, the content of FA contributes to crobiota fermentation rate and the amount of produced SCFAs
this property as well. Bound FA demonstrates a cross-linking effect (Table 1). Vardakou et al. (2008) observed an increase in the prebiotic
which improves the gel-forming ability of AXs (Döring et al., 2016). index with AXs treated by xylanase compared to AXs without xylanase
Aside from AX’s own structural features, a higher concentration of AXs treatment. Similarly, Geraylou et al. (2013) showed that a higher DP of
also increases the viscosity. AXOS supplementation exerted decreased impacts on SCFA concentra-
tion compared with lower DP. More researches also showed that higher
4. The interaction between AXs and gut microbiota DP reduced the fermentation rate of AXs and AXOS in human colon
(Audrey et al., 2016; Gemen et al., 2011; Pollet et al., 2012). However,
4.1. The pathway to produce short chain fatty acids (SCFAs) supplementation of water-extractable high MW AXs restore the number
of bacteria that are lowered upon high fat feeding (Neyrinck et al.,
Due to the nature that AXs can’t be degraded by mammalian en- 2011). Van den Abbeele et al. (2011) demonstrated that long-chain AXs
zymes, they end up in colon and are degraded by gut microbiota. Ndeh with high DP significantly stimulated the production of health-pro-
and Gilbert (2018) showed that gut microbiota degraded AXs into moting metabolites by specific microbes in the proximal regions of the
SCFAs and other products (Fig. 2). The fermentation of AXs was mainly colon in humanized rats. Damen et al. (2011) reported that feeding rat
through the interaction with gut microbiota and catalyzed by backbone with a higher DP water unextractable AXs diet compared with water
xylan depolymerizing species, especially Bacteroidetes. It has been re- extractable AXs, the number of butyrate-producing bacteria like Rose-
ported that AXs were possessed by starch utilization system (Sus)-like buria and E. rectale spp. were increased, which led to increased butyrate
system encoded by polysaccharide utilization loci (PUL) from the level in the rat’s cecum. These results suggested that AXs with high MW
phylum Bacteroidetes (Koropatkin, Cameron, & Martens, 2012). The sus- or DP might have positive effects on the fermentation rate as well as
like system contained a set of polysaccharide binding-proteins (SusD SCFA production, thus the influence of DP of AXs requires further in-
homologue; SusDH), glycolytic enzymes that hydrolyze polysaccharides vestigation. Compared with rice AXs, maize AXs were shown to be less
into oligosaccharides, and TonB-dependent transporters that trans- branched. The bacteria were required to remove the arabinose before
porting these oligosaccharides into the periplasm (SusC homologue; being able to utilize xylose of rice AXs, but the maize AXs with com-
SusCH) (Dodd, Mackie, & Cann, 2011; Martens, Koropatkin, Smith, & paratively less branched resulted in easy access for bacteria to reach
Gordon, 2011; Ndeh & Gilbert, 2018). Glycoside hydrolases on Bac- xylose and generate higher SCFAs production (Rose, Patterson, &
teroidetes surface degraded AXs into arabinoxylooligosaccharides Hamaker, 2010). Therefore, highly branched oligosaccharides are
(AXOS). AXOS then were transported into the cell periplasm by SusCH/ poorly fermented (Rose et al., 2010). Meanwhile, fermentation patterns
SusDH pairs (Glenwright et al., 2017; Ndeh & Gilbert, 2018). The de- of maize, rice and wheat AXs were different. Maize and rice AXs were
polymerization pathway of AXOS in cell periplasm of Bacteroidetes, degraded by a debranching mechanism while wheat AXs were firstly

538
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

Fig. 2. Depiction of the possibly process of


diet colon the interaction between gut microbiota and
arabinoxylans (AXs) and its hydrolysis pro-
Firmicutes and Actinobacteria ducts arabinoxylooligosaccharides (AXOS),
to produce short chain fatty acids (SCFAs).
Bacteroidetes AX XOS The AXs fermentation pattern of
AX AX AX Bacteroidetes is through starch utilization
system (Sus)-like system encoded by poly-
AX saccharide utilization loci (PUL). AXs may
X degrade by glycoside hydrolases (possibly
AXOS AXOS AXOS endoxylanase) on the surface into AXOS.
A SCFA AXOS may be transported into the cell
periplasm by SusCH/SusDH pairs. The de-
polymerization pathway of AXOs in cell
AXOS AXOS periplasm of Bacteroidetes is unclear. Beside
SCFA
Bacteroidetes, Roseburia from Actinobacteria
has the ability to degrade AX as well. The
AXs and Bacteroidetes downstream metabo-
lism can be transported into Firmicutes or
Actinobacteria through four kinds of prob-
starch utilization system (Sus)-like system able transporters and depolymerized by
ATP-binding cassettes transporters carbohydrate-Active Enzymes which en-
coded by gram positive PULs. AXs are in-
Cation symporter family transporters itially degrade by endoxylanase into AXOS
Major facilitator superfamily transporters followed by AXOS linkage breakdown by
arabinofuranosidase and xylosidase into
PEP-phosphotransferase system transporters arabinose (A) and xylose (X). Secondly, the
produced xylose (X) and arabinose (A) can
endoxylanase be used by some species of Bifidobacterium
and Roseburia to acquire SCFAs.
arabinofuranosidase
xylosidase

degraded by bacteria in its unsubstituted region followed by their substituted by the relatively uniform arabinose. The complexity of the
arabinose side chains being metabolized, indicating a two-phase me- AXs molecules was shown to strongly influence the fermentation rate
tabolism (Banuls et al., 2016). Thus, DS is an important structural factor (Chen et al., 2017). This opinion was also confirmed by Rumpagaporn’s
associated with the fermentation, which influences not only SCFA observation that a slow fermentation rate of AXs was associated with
production, but also fermentation mechanisms. Chen et al. (2017) in- more branches, single xylose units, or the unusual trisaccharide branch
vestigated that sorghum bran AXs were fermented more rapidly com- chains in AXs (Rumpagaporn et al., 2015). Mendis, Martens, and
pared to corn AXs due to the different branched structure. Both of Simsek (2018) investigated the fine structure of AXOS and XOS influ-
sorghum bran AXs and corn AXs had similar molecular size but corn ence the growth of Bacteroides species in the intestine, which suggested
AXs had a more complex composition of sugars and linkage patterns that 23– α-L-arabinofuranosyl-xylotriose could elevate the expression of
compared to sorghum bran AXs in which the xylan backbone was small xylan PUL gene while 23, 33 -di- α-L-arabinofuranosyl-xylotriose

Table 1
Effects on interaction with gut microbiota.
Molecular structure Effect on interaction with gut microbiota In vivo/in vitro Reference

AXs (avDP = 60, avDS = 0.70) Stimulate bacterial species with butyrate In vivo, rats (Van den Abbeele et al.,
producing ability and bifidobacteria. Reduced 2011)
caecal abundances of Akkermansia muciniphila
AXs (avDP = 284, avDS = 0.59) Increased the number of butyrate producing In vivo, rats (Damen et al., 2011)
bacteria like Roseburia and E. rectale spp.
AXs from wheat bran (Ara/Xyl ratio, 1.14; contained a complex AXs from wheat bran fermented slowly compared In vitro, human fecal (Rumpagaporn et al.,
trisaccharide side chain; high amounts of branches with single to sorghum AXs due to the branched structure fermentation analyse 2015)
xylose units); sorghum AXs (Ara/Xyl ratio, 1.08; contained the differences.
highest degree single arabinosebranches)
AXs from maize bran (Ara/Xyl ratio, 0.52) AXs from rice bran (Ara/ Less branched maize AXs resulted in the highest In vitro, digestion and (Rose et al., 2010)
Xyl ratio, 1.12) SCFA production. fermentation analyse
Sorghum bran AXs (contained uniform arabinose substitution); corn Sorghum bran fermented rapidly compared to corn In vitro, human fecal (Chen et al., 2017)
AXs (complex array of sugars and linkage patterns) AXs due to the branched structure differences fermentation analyse
AXOS1 (avDP = 3); AXOS2 (avDP = 12) High avDP AXOS has slower fermentation rate In vitro, human fecal (Snelders et al., 2014)
compared with AXOS with low avDP fermentation analyse
AXOS1 (high bound FA content); AXOS2 (high free FA content); Both bound and free FA inhibited AXOS In vitro, human fecal (Snelders et al., 2014)
AXOS3 (low FA content) fermentation fermentation analyse
AXOS1 (avDP = 32, avDS = 0.30); AXOS2 (avDP = 3, avDS = 0.25) AXOS (avDP = 32, avDS = 0.30) has higher In vivo, fish (Geraylou et al., 2013)
impact on SCFAs production and more pronounced
effects on stimulation of Lactobacillaceae

avDP, average degree of polymerization; avDS, average degree of substitution; Ara/Xyl Ratio, arabinose/xylose ratio; AXs, arabinoxylans; AXOS, arabinox-
ylooligosaccharides; FA, ferulic acid; SCFAs, short chain fatty acids.

539
Z. Chen et al.

Table 2
Effect on metabolism of AXs with different structural features.
Sources Molecular structure Effect on metabolism In vivo/in vitro Reference

Effect on lipid metabolism


Wheat bran Ara/Xyl ratio, 0.58 Increase postprandial fat utilization In vivo, mice (Hosoda et al., 2017)
Wheat bran MW, 602 kDa; Ara/Xyl ratio, 0.9 Increase fatty acid oxidation; reduce plasma cholesterol; increase ileal viscosity In vivo, diabetic fatty rats (Hartvigsen et al., 2013)
Wheat bran Ara/Xyl ratio, 0.47 Decrease circulating bile acids In vitro, 13C NMR and SAXS experiments (Gunness, Flanagan, et al., 2016; Gunness,
Williams et al., 2016)
Wheat bran MW, 63.2–3470 kDa Reduce plasma cholesterol In vivo, hypercholesterolemic hamsters (Tong et al., 2014)
Wheat bran Ara/Xyl ratio, 0.7 Decrease lipid synthesis in adipose tissue In vivo, obese mice (Neyrinck et al., 2011)
Finger millet Ara/Xyl ratio, 0.78 Decrease lipid synthesis in liver In vivo, mice (Sarma et al., 2018)
Rye Ara/Xyl ratio 0.45 Decrease circulating bile acids In vitro (Gunness, Flanagan, et al., 2016; Gunness,
Williams et al., 2016)
Corn bran avDS = 0.64 Reduced cholesterol uptake. Decreased serum cholesterol levels. Inhibited In vivo, rats (Lopez et al., 1999)
cholesterol accumulation in liver.
Wheat bran AXOS (contained FA) Inhibited lipid peroxidation in serum, liver and testes. In vivo, diabetic rats (Shiyi Ou et al., 2007)

540
Wheat bran AXOS Reduced body weight, fat mass and plasma leptin levels. In vivo, mice (Suriano et al., 2017)

Effect on glucose metabolism


Wheat bran MW, 602 kDa; Ara/Xyl ratio, 0.9 Increase glycolysis; increase ileal viscosity; attenuate plasma glucose In vivo, diabetic fatty rats (Christensen et al., 2013)
Wheat bran Ara/Xyl ratio, 1.23; contained FA Inhibit glucose transporter 2 and α-glucosidase In vitro (Malunga et al., 2016)
Destarched bran Cross-linked AXs Remained viscous in theintestinal and lowered the postprandial blood glucose In vivo, rats (Vogel, Gallaher, & Bunzel, 2012)
level.
Wheat flour avDS = 0.8 Reduced postprandial serum glucose, insulin and blood plasma ghrelin. In vivo, human (Garcia et al., 2007; Garcia et al., 2006)
Decreased fasting serum glucose
Wheat flour avDS = 0.8 Lowered postprandial serum insulin; elevated postprandial blood plasma ghrelin. In vivo, human (Möhlig et al., 2005)
Wheat flour avDS = 0.66 Decreased fasting serum glucose levels. Reduced serum glucose levels and insulin In vivo, human (Lu, Walker, Muir, & O'Dea, 2004)
concentration 2 h after oral glucose intake.
Wheat bran AXOS (Ara/Xyl ratio, 0.2–0.3; MW, Reduced metabolic endotoxemia thereby inhibiting insulin resistance In vivo, diabetic rats (Neyrinck et al., 2012)
below 0.59 Da)
Wheat bran AXOS (contained FA) Inhibited weight loss of diabetic; down regulated serum glucose level. In vivo, diabetic rats (Shiyi Ou et al., 2007)

avDS, average degree of substitution; Ara/Xyl Ratio, arabinose/xylose ratio; MW, molecular weight; AXs, arabinoxylans; AXOS, arabinoxylooligosaccharides; FA, ferulic acid.
Journal of Functional Foods 54 (2019) 536–551
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

lowered the expression. Furthermore, the presence of feruloylated and The physicochemical properties and the interaction with gut mi-
diferuloylated arabinose substituents reduced the fermentability of AXs crobiota can be influenced by AXs’ structural features. Further more,
and AXOS, because bound FA probably hindered enzyme activity the physicochemical properties of AXs performed in vivo and the
sterically, but bound FA did not impact the SCFA ratio (Hopkins et al., change in the composition of gut microbiota may be the mechanisms to
2003; Snelders, Dornez, Delcour, & Courtin, 2013; Snelders et al., explain the physiological functions of AXs. Nowaday, the physiological
2014). functions of AXs are receiving growing attention. We analyzed the
Above all, it can be concluded that the DP, DS, chemical composi- mechanisms of AXs to perform health benefits and discussion the re-
tion and linkage-type of the branches, all together, affect the rate of lationship between AXs structure and physiological functions.
fermentation of gut microbiota. AXs with lower molecule weight, lower
DS, less FA content and simple linkage and sugar pattern will lead to a 5. Physiological functions of AXs
higher fermentation rate and generate more SCFAs. It is widely known
that the fermentation product SCFAs exert beneficial physiological 5.1. The effect of AXs on lipid metabolism
functions in human. Therefore, it is possible that the structural of AXs
influenced AXs’ health effects through changing the fermentation rate Several studies investigate the physiological function in lipid me-
and SCFAs production. tabolism of AXs with different structures (Table 2). AXs are able to
increase lipid excretion in feces of mice due to its fat binding capability
4.3. The change of gut microbiota (Neyrinck et al., 2011; Suriano et al., 2017). The fat binding capability
can be one of the mechanisms explaining the decline in ectopic lipid
The interaction between AXs and gut microbiota takes place in the accumulation in the liver tissue, which can attribute to the decrease in
intestine, the abundance and the composition of intestinal flora are dietary nonesterified fatty acids that redistributed from the adipose
likely to be changed after AXs supplementation. Several research tissue to the liver (Suriano et al., 2017). After the experiment in rats,
groups observed that the numbers of Roseburia intestinalis, Eubacterium Lopez et al. (1999) investigated that soluble corn bran AXs declined the
rectale, Anaerostipes caccae, Clostridium clusters XIVa, and some other uptake of cholesterol from a cholesterol-rich diet, leading to the de-
saccharolytic colon bacteria, such as Bacteroides spp. as well as creased in total cholesterol as well as low-density lipoprotein (LDL)-
Lactobacillus spp. increased in rats upon AXs supplementation (Damen cholesterol in the plasma. Additionally, the absorption of lipids in-
et al., 2011; Van den Abbeele et al., 2011). However, AXs have been cluding long chain fatty acid-like linoleic acid and cholesterol was
shown to be rarely bifidogenic (Broekaert et al., 2011). At the same slowed down by AXs in diabetic fatty rats, possibly through increasing
time, the studies on AXOS (the hydrolytic degradation products of AXs) the viscosity of the surrounding intestinal digesta (Gunness, Flanagan,
showed that after feeding fish (juvenile Siberian sturgeon) with a diet Shelat, Gilbert, & Gidley, 2012; Hartvigsen et al., 2013). After the ki-
containing 2% AXOS, the amount of Eubacteriaceae, Streptococcaceae, netic analysis in vitro, it has been observed that increased viscosity of
Lactobacillaceae, Lactococcus lactis, and Bacillaceae increased (Geraylou the surrounding intestinal bolus led to the inhibition of the contents
et al., 2013). Likewise, it has been demonstrated that the levels of bi- mixing with the unstirred water layer thereby elevated the effective
fidobacteria, lactobacilli, and Eubacterium rectale were increased, while thickness of the unstirred water layer (Gunness et al., 2012). This thick
that of Bacteroides was reduced upon AXOS supplementation in vitro viscous unstirred water layer was composed of a diffusion barrier for
during fermentation by human gut derived microbiota (Audrey et al., the absorption and the augment in its thickness led to lower lipid ab-
2016; Sanchez et al., 2009; Snelders et al., 2014). Using the experi- sorption (Gunness et al., 2012). It was also believed that AXs lowered
mental setup constituted two identical units of the SHIME, Grootaert the speed of triglycerides digestion as well as free fatty acids assim-
et al. (2009) investigated that AXOS degradation in the colon was in- ilation through decreasing circulating bile acids (BA), leading to re-
dicated to be compartment-specific depending on their average DP. duced blood triglycerides level in pigs (Gunness, Williams et al., 2016).
Those with an average DP lower than 15 were almost completely uti- AXs interacted with BA through dynamic molecular interaction or
lized in the ascending and transverse colon compartments in the si- polysaccharide aggregates entrapment in vitro (Gunness, Flanagan,
mulator of human intestinal (Sanchez et al., 2009). As for the AXOS Mata, Gilbert, & Gidley, 2016). BA is an important substance con-
with an average DP of 29, the abundance of bifidobacteria in the as- tributing to dissolve and transit hydrophobic entities (Shelat et al.,
cending colon, lactobacilli in both ascending and transverse colons was 2010). Besides, the arabinose residues of AX’s side chain contribute to
augmented (Sanchez et al., 2009). It seems that AXs and AXOS exert an reducing BA absorption, due to its ability to infiltrate into hydrophilic
inconsistent impact on the abundance and the composition of gut mi- head of BA micelles (Gunness, Flanagan, et al., 2016). Nie et al. (2018)
crobiota, especially in Bifidobacterium and Bacteroides. This inconsistent showed that AXs treatment reduced the amount of acetoacetate,
impact is likely attributed to the different structure of AXs and AXOS. acetone, and choline in type 2 diabetic rats thereby played a role in
Bifidobacterium species mainly produce xylosidases and arabinofur- lipid metabolism. The fatty acid metabolism in the liver mitochondria
anosidases while Bacteroides mainly produce endoxylanase (Broekaert produces acetoacetate and acetone, two main products in ketone body’s
et al., 2011; Chassard, Goumy, Leclerc, Del'Homme, & Bernalier- metabolism.
Donadille, 2010; Lambertusam & Alphonsgj, 2008; Zeng, Xue, Peng, & Besides the impact on digestion of lipid, the gene expression of lipid
Shao, 2007). For the AXs with high DP, they tend to be degraded by is beneficially regulated by AXs supplementation in the liver. The ex-
endoxylanase which stimulated the growth of Bacteroides. While for the periments in mice was observed that AXs have been shown to decrease
AXOS with the low DP, it is easier to be accessed by xylosidases and lipogenic gene expression such as fatty acid synthase, acetyl-CoA car-
arabinofuranosidases to produce xylose and arabinose, and conse- boxylase, sterol regulatory element-binding protein-1, and upregulate
quently the abundance of Bifidobacterium. increases. Furthermore, long- fatty acid oxidation gene expression, for instance, acyl-CoA oxidase-1,
chain AXs supplementation has been shown to increase caecal mucin Cyp4a10, Cyp4a14 and cytochrome p450, suggesting AXs reduced li-
level by lowering caecal abundances of the mucin-degrading Akker- pogenesis and increased β-oxidation thereby decreased lipid accumu-
mansia muciniphila (Van den Abbeele et al., 2011). lation in the liver and decreased triglycerides in the serum and in the
AXOS supplementation inhibited Salmonella colonization in broiler liver (Kieffer et al., 2016; Sarma et al., 2018). AXs also have been
chickens (Eeckhaut et al., 2008). Salmonella is a pathogenic bacteria shown to increase expression of cholesterol 7- alpha-hydroxylase to
and is harmful to human health. Likewise, since AXOS can stimulate accelerate cholesterol catabolism and BA synthesis in hamsters (Tong
acetate and propionate production thereby lower the pH in intestinal, it et al., 2014). Likewise, AXs decrease cholesterol synthetic gene ex-
also inhibits the growth of E. coli in vitro (Cherrington, Hinton, Pearson, pression such as 5-hydroxy-3-methlgutaryl-coenzyme A reductase and
& Chopra, 2010). 3-hydroxy-3-methylgulutaryl-CoA synthase due to elevated propionate,

541
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

to exert hypocholesterolemic effect in vivo (Chen et al., 2018; Tong plasma from portal–arterial catheterised pigs was conducted, the results
et al., 2014). Furthermore, in high-fat diet induced rats, liver damage in shown that AXs supplementation caused a reduction of glucose uptake,
rats can be relieved by AXs supplementation due to the regulation of leading to a lower concentration of lysophosphatidylcholine (Nielsen,
liver cell apoptosis and a decrease of the lipid hydroperoxides (Chen Hedemann, Laerke, Jorgensen, & Bach Knudsen, 2014). Lysopho-
et al., 2018). sphatidylcholine is elevated in the acquired obesity and is the main
The gene expression in adipose tissue is modulated by AXs supple- plasma lipid component of oxidized LDL, suggesting that high lyso-
mentation as well. Sarma et al. (2018) showed that AXs supplementa- phosphatidylcholine concentration likely result in chronic inflamma-
tion regulated peroxisomeproliferator-activated receptor gamma, tion as well as atherosclerosis (Nielsen et al., 2014).
CCAAT/enhancer-binding protein alpha and delta-like-1 of mice, to The structure of AXs influences AXs’ ability of lipid metabolism
modulate transformation rate of preadipocytes into mature adipocytes. modulation. Viscosity of AXs plays an important role in fat binding
The transformation rate was correlated with adipocyte hyperplasia. At capacity, lipid and BA absorption. Thus, the structural features of AXs
the meantime, AXs supplementation may also decrease fatty acid syn- may change the viscosity to exerts effect on AXs’ ability of lipid meta-
thase and downregulates peroxisome proliferator-activated receptor bolism modulation. Moreover, at the same time, SCFAs are of great
alpha, elevates acyl-CoA oxidase-1 expression through stimulating significance in modulating lipid metabolism. The structural features of
propionate production in vivo, suggesting that it decreases lipid AXs can change the interaction with gut micromiota of AXs thereby
synthesis and increases lipid breakdown (Heimann, Nyman, & influence the production of SCFAs. However, although several animals
Degerman, 2015; Hosseini, Grootaert, Verstraete, & Van de Wiele, experiments demonstrated that AXs supplementation change the gene
2011; Neyrinck et al., 2011). Consequently, adipocyte hypertrophy is expression in liver or adipose tissue, the mechanism of them are still
decreased in visceral white adipose tissue. AXs can modulate adipocyte unclear. Further researches may need to focus on the bioactive fraction
hypertrophy and adipocyte hyperplasia which are the two factors of AXs leading to the change in gene expression, and figure out whether
contributed to adipose tissue mass augment (Jiang, Jo, & Graff, 2012). gut microbiota are involve in the mechanism.
In white adipose tissue, fasting-induced adipose factor expression is
downregulated after AXs supplementation in the high fat diet. Using
transgenic mice with over-expressed fasting-induced adipose factor, 5.2. The effect of AXs on glucose metabolism
Neyrinck et al. (2011) observed that fasting-induced adipose factor
could elevate total cholesterol as well as high-density lipoprotein cho- The effects of various soluble AXs on glucose metabolism have been
lesterol levels. Moreover, AXs can inhibit lipolysis and de novo lipo- documented (Table 2). The soluble AXs likely delay gastric emptying,
genesis in adipose tissue through stimulating propionate production in slow small intestinal transit, reduce the rate of glucose diffusion, de-
vivo, which plays an important role in preventing ectopic fat deposition crease the availability of digestive enzymes, and mediate intestinal
and lipotoxicity (Heimann et al., 2015). mobility, all contribute to lower the absorption of glucose and an at-
The interaction of AXs with gut microbiota influences lipid meta- tenuated postprandial glycemic response (Dhital et al., 2014; Giulia
bolism. It has been reported that in rats which were supplied with a Falchi, Grecchi, Muggia, Palladini, & Perlini, 2016; Juvonen et al.,
high-fat diet and the obese adults had a high endogenous acetate pro- 2009; Vangsøe et al., 2016). Additionally, the gel-forming property of
duction, a probable biomarker leading to metabolic syndrome (Chen soluble AXs is able to slow down the protein degradation and absorp-
et al., 2018). Long-chain AXs were able to decrease acetate production tion. Thus, the level of amino acid is decreased and lead to an atte-
through transforming acetate into health-promoting propionate and nuated insulin response (Christensen et al., 2013). Viscosity, or gel-
butyrate in rats or mice (Berger et al., 2014; Fadel et al., 2017; Van den forming capacity, is a physicochemical property associated with AXs,
Abbeele et al., 2011). Heimann et al. (2015) showed that propionate which is one of the mechanisms of AXs’ ability of affecting short-term
and butyrate changed the gene expression in the adipose tissue of rats postprandial glucose level2. The effect, however, may be offset by
to decrease lipid synthesis and increase lipid breakdown. Likewise, they strong intestinal peristalsis, indicating that it would not be the principal
prevented ectopic fat deposition and lipotoxicity through inhibition of factor of reducing the digestion rate in the intestine (Dhital et al., 2014;
lipolysis and de novo lipogenesis. AXs supplementation increases Ro- Gemen et al., 2011). The high or low viscosity of AXs relates to different
seburia spp while AXOS supplementation elevates both Bifidobacteria physiological functions. It has been reported that high viscosity exerted
and Roseburia spp. in mice (Druart et al., 2013; Neyrinck et al., 2011). beneficial effects on glycemia and insulinemia due to slower gastric
Bifidobacteria and Roseburia spp are able to metabolize linoleic acid into emptying rate (Juvonen et al., 2009). A higher nutrient-mediated GLP-
conjugated linoleic acid isomers (Druart et al., 2013; Neyrinck et al., 1, however, was triggered by a reduction of viscosity, which facilitated
2011). At the same time, AXs supplementation elevated linoleic acid’s insulin synthesis as well as evoked cholecystokinin and peptide YY re-
availability for the gut microbiota in the lower part of the gut, due to its sponse (Hartvigsen et al., 2014; Juvonen et al., 2009). Therefore, the
fat binding capacity (Druart et al., 2013). Both properties contribute to ghrelin release was inhibited with the improvement in a sense of satiety
a higher production of conjugated linoleic acid (Druart et al., 2013; (Hartvigsen et al., 2014; Juvonen et al., 2009). It can be concluded that
Neyrinck et al., 2011). It has been reported that conjugated linoleic acid high viscous AXs are apt to reduce postprandial glucose level than low
can inhibit the enzymes related to mammary lipid synthesis (Bauman, viscous AXs, but are more difficult to improve a sense of satiety and
Harvatine, & Lock, 2011). Neyrinck et al. (2018) believed that AXOS may lead to higher energy intake for a healthy subject. The molecular
supplementation exerted bifidogenic effect associated with a decrease structures of AXs that affect the viscosity properties including MW,
in body weight, reduced fat mass, and attenuated plasma leptin levels in arabinose: xylose ratio and the degree of branching (Christensen et al.,
mice. Interestingly, such effects did not depend on caloric intake or the 2013; Gemen et al., 2011). Furthermore, the food processing method(s),
regulation of peptides affecting appetite such as ghrelin or glucagon- as well as the combination of foods and drinks during a meal will also
like peptide 1 (GLP-1) (Neyrinck et al., 2018). result in different AXs viscosity (Gemen et al., 2011; Pantophlet et al.,
The ability of AXs to modulate lipid metabolism is related to glucose
metabolism. A diet containing AXs causes a reduction of the post- 2
In rats, a diet with low viscosity AXs did not have ability to decrease
prandial blood glucose-dependent insulinotropic polypeptide response
postprandial glucose, however, cross-linking of the AXs remarkably elevated
in mice, leading to an increase in postprandial energy catabolism and viscosity and attenuated the glucose response (Vogel et al., 2012).In a research
prevention of obesity (Hosoda et al., 2017; Neyrinck et al., 2018). conducted by Wood et al. (1994), nine human subjects consumed liquid meals
Glucose-dependent insulinotropic polypeptide is a gastrointestinal in- contained glucose and different concentration of oat gum. It was observed that
cretin hormone, which promotes pancreatic β cells to secrete insulin. the plasma glucose concentration reduced when the subjects consumed high
Likewise, the liquid chromatography–MS metabolomics analysis of viscosity of oat gum.

542
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

avDP, average degree of polymerization; avDS, average degree of substitution; Ara/Xyl Ratio, arabinose/xylose ratio; MW, molecular weight; AX, arabinoxylan; AXOS, arabinoxylooligosaccharides; FA, ferulic acid; PAP,
2017).

(Samuelsen et al., 2011)


(Cholujova et al., 2013)

(Neyrinck et al., 2012)


(Niewold et al., 2012)
By inhibiting mammalian intestinal α-glucosidase (sucrase and

(Kamiya et al., 2014)

(Zhang et al., 2004)


(Zheng et al., 2012)
(Zhou et al., 2010)
(Zhou et al., 2010)
(Cao et al., 2011) maltase) and reducing glucose transporter 2 activities, feruloylated

(Das et al., 2013)


arabinoxylan mono/oligosaccharide shows an antihyperglycemic ef-
fect. The inhibitory activity is attributed to FA moiety, an effective part
Reference

of feruloyl arabinoxylanmono/oligosaccharide (Malunga, Eck, & Beta,


2016). In the diabetic fatty rats fed with AXs contained diet, the ex-
pression of fat-binding protein 2 was upregulated, consistent with the
In vivo, S180tumor-bearing mice

higher insulin level. Fat-binding protein 2 was thought to be related to


improvement of insulin sensitivity (Hartvigsen et al., 2013). Using type
2 diabetic rats, Nie et al. (2018) observed reduction in pyruvate
vivo, broiler chicks

vivo, diabetic rats


through high-dose Plantago asiatica L. AXs supplementation. Pyruvate is
an important product in glycolysis, suggesting that AXs supplementa-
In vivo/in vitro

In vivo, human

In vivo, human

vivo, piglet

tion is able to attenuate glycolysis in type 2 diabetic rats from the


In vivo, rats
In vivo, rats

In vivo, rats

general view (Nie et al., 2018). However, in adipose tissue, an aug-


vitro
vitro

mented level of insulin followed by up-regulated adipose glycer-


aldehyde-3-phosphate dehydrogenase expression was observed in the
In
In
In
In
In

diabetic fatty rats fed with AXs contained diet. Glyceraldehyde-3-


Stimulated immature dendritic cell maturation. Augmented NK cytolytic activity
had the ability to reduce the IL-8 concentration, exerted a protective effect on

phosphate dehydrogenase participates in glycolysis, indicating that AXs


Strengthens T and B cell functions, elevates lymphocyte ratio, decreases the
Stimulated macrophage phagocytosis and delayed hypersensitivity reaction

promoted glycolysis in the adipose tissue (Hartvigsen et al., 2013),


specific mechanism need further exploring. In addition, AXs supple-
increase in antibacterial protein PAP to against acute ETEC challenge
Improve NK cells cytotoxic activity and activation of T and B cells

mentation restores tricarboxylic acid cycle intermediates in urine (2-


Induced expression of IL-10 to perform anti-inflammatory effect.

oxoglutarate, succinate, and malate), which normally is elevated in


type 2 diabetic rats, indicating that AXs have the ability to inhibit
Strong splenocyte, thymocyte, and macrophage activations
exerted effects on innate and acquired immune responses

diabetes triggered augment of tricarboxylic acid cycle (Nie et al., 2018).


Kieffer et al. (2016) reported that enzyme-treated wheat bran-fed mice
(AXs and AXOS were the main components of diet) showed an upre-
gulated expression of phosphoenolpyruvate carboxykinase 1 and glu-
Elevated macrophages activation in vitro

cose-6-phosphatase. Phosphoenolpyruvate carboxykinase 1 is a speed-


limiting enzyme for gluconeogenesis while glucose-6-phosphatase is
related to gluconeogenesis and glycogenolysis through gut-derived
Low immunological responses

signals. Thus, enzyme-treated wheat bran supplementation modulated


Effect on the immune system

both gluconeogenesis and glycogenolysis which might play important


roles in glucose homeostasis in the liver (Kieffer et al., 2016).
AXs and AXO supplementation cause an important bifidogenic effect
neutrophil ratio

by promoting L cells’ GLP-1 and glucagon-like peptide 2 (GLP-2) pro-


duction (Neyrinck et al., 2012). GLP-1 is an intestinal peptide capable
hepatitis

of regulating food intake and glucose homeostasis. The augment of


GLP-2 promotes the improvement of intestinal barrier function to pre-
vent metabolism endotoxemia through increasing the expression of
Low molecular weight fraction of MGN-3 (MW lower than

tightly bound proteins (Broekaert et al., 2011; Neyrinck et al., 2012).


Metabolism endotoxemia is one of the triggering factors leading to the
AXOS (Ara/Xyl ratio, 0.2–0.3; MW, below 0.59 Da)

development of insulin resistance (Geurts, Neyrinck, Delzenne, Knauf,


Ara/Xyl ratio, 0.56; MW, 32.5 kDa; contained FA

& Cani, 2014; Neyrinck et al., 2012). In addition, consumption of the


MGN-3 (Ara/Xyl ratio, 0.5; MW, 30–50 kDa)

MGN-3 (Ara/Xyl ratio, 0.5; MW, 30–50 kDa)

AXs-rich diet increased SCFAs absorption and, in particular, butyrate


Effect on the immune system of AXs with different structural features.

absorption (Ingerslev, Theil, Hedemann, Lærke, & Bach Knudsen,


AXOS (avDP = 5, Ara/Xyl ratio, 0.21)

2014). Higher butyric acid uptake elevates glucose intake in adipocytes


Ara/Xyl ratio, 0.83; MW, 351 kDa

stimulated by insulin and particularly improves peripheral insulin


sensitivity by affecting adipose tissue as well as circulating non-
pancreatitis associated protein; ETEC, Escherichia coli.

esterified fatty acids. Insulin sensitivity may increase glucose tolerance


in an overnight perspective (Boll et al., 2016; Heimann et al., 2015;
Molecular structure

Ingerslev et al., 2014). Propionate stimulates the production of GLP-1 in


MW, 351 kDa

MW, 175 kDa


MW, 156 kDa
MW, 288 kDa

the gut and improves satiety by promoting insulin synthesis which re-
sults in inhibition of ghrelin release (Broekaert et al., 2011). Likewise,
400 Da)

propionic acid or, to some extent, butyric acid, can affect basal lipolysis,
which may exert some positive effects on glucose homeostasis in me-
tabolic disorders (Heimann et al., 2015). AXs also increase absolute
Green leaves of Litsea glutinosa

concentration of acetic acid, inhibit insulin-stimulated glucose uptake,


and suppress insulin signaling by inhibiting the protein kinase B in 3T3-
L1 adipocytes and primary adipocytes cells of mice (Heimann et al.,
2015; Kimura et al., 2013; Van den Abbeele et al., 2011). It seems that
small intestinal microbiome is able to break down AXs into arabinose
banana peel
Wheat bran
Wheat bran
Wheat bran

Wheat bran
Wheat bran

and xylose (Pantophlet et al., 2017). Arabinose itself reduces the starch-
Rice bran

Rice bran
Rice bran
Sources

derived glucose production via suppressing maltase activity in the small


Table 3

Barley

intestine (Jurgoński, Krotkiewski, Juśkiewicz, & Billing-Marczak,


2015).

543
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

Same as the effect of AXs on lipid metabolism, it has been observed reduce the IL-8 concentration and its mRNA expression level, which
that AXs change gene expression related to glucose metabolism, how- exerted a protective effect on D-GalN-induced hepatitis, indicating that
ever, the mechanisms are still unclear. It is possible that the bioactivate low MW compounds in MGN-3 seemed to be effective (Zheng et al.,
fractions of AXs change the gene expression or AXs change the com- 2012).
position of gut microbiota thereby modulating the gene expression. AXs supplementation can lower pH of the cecal (Chen et al., 2015).
Likewise, the ability of AXs to decrease glucose level in plasma may Low pH level has been shown to inhibit the number of pathogenic
lead to body adjusted function. This may also be one of the mechanisms bacteria, thus, is considered to be beneficial (Chen et al., 2015). It was
to explain the change in gene expression. reported that the level of Lactobacillus plantarum was elevated in the
pigs who were fed with AXOS (Niewold, Schroyen, Geens, Verhelst, &
5.3. The effect of AXs on the immune system Courtin, 2012). The increase of Lactobacillus plantarum was related to
the increase of pancreatitis-associated protein, an antimicrobial protein,
The immune-enhancing function of AXs have been studied which was important in innate immune responses in acute E. coli
(Table 3). AXs from wheat bran, 3.517 × 105 Da MW, enhance the challenge (Niewold et al., 2012). Similarly, the activity of the intestinal
macrophage phagocytosis of chicken red blood cells and improve the alkaline phosphatase increased which was related to the stimulation of
cytotoxic activity of NK cells against derived tumor cells. Meanwhile, lactobacillus in the distal colonic mucosa of the AXs-fed mice. Intestinal
AXs remarkably elevate concanavalin A level and promote lipopoly- alkaline phosphatase was able to detoxify lipopolysaccharide and pre-
saccharide-stimulated splenocyte proliferation in S180 tumor-bearing vent bacterial invasion across the gut mucosal barrier (Chen et al.,
mice. They also dose-dependently increase IL-2 secretion in the blood 2015). AXs could reduce the adhesion of E. coli through competitive
serum. All these effects contributed to the activation of T and B cells rejection via stimulated Latic bacteria (Hermes, Manzanilla, Martin-
(Cao et al., 2011). It has been reported that AXs increased total im- Orue, Perez, & Klasing, 2011). The results demonstrate that AXs sup-
munoglobulins (Igs), immunoglobulin (Ig)G and IgM anti-sheep red plementation promotes making an unfavourable environment towards
blood cells antibody titers in chickens (Akhtar et al., 2012). A higher pathogen (Fu et al., 2011), which prevents further infection of patho-
level of anti- sheep red blood cells IgM antibody titers has been asso- gens and therefore enhances the immune response. An additional study
ciated with attenuated humoral immunity, possibly due to that AXs shown that rice bran could increase the colonization of Lactobacillus
were able to strength physiological and infectious insults resistance which might due to elevated mucosal IgA response (Henderson, Kumar,
(Akhtar et al., 2012). Likewise, AXs from corn hull, 5.06 × 105 Da MW Barnett, Dow, & Ryan, 2012). The exact effect of AXs supplementation
and contained D-galactose and α-D-glucuronic acid, elevate NK cell on IgA response, however, is still not clear. Furthermore, several studies
activity and spleen cells’ TNF-α secretion in the presence of con- showed that AXs supplementation was beneficial to increase and
canavalin A and lipopolysaccharide leading to a lower tumor growth modulate mucus production (Chen et al., 2015). In the mean time, it is
rate during the early stages after tumor transplantation (Ogawa et al., believed that the decrease in mucus thickness related to microbiota
2005). Similarly, AXs from banana peel hemicellulose, 2.88 × 105 Da activity would increase pathogen susceptibility (Desai et al., 2016). A
MW and contained galactose, elevate macrophages activation in vitro fibrous diet containing AXs protects the mucus barrier and protein
(Zhang et al., 2004). Zhou et al. (2010) investigated the immune-en- barrier factors through increasing goblet cells and stimulating the
hancing function of alkaline extracted AXs (AXA) (MW = 3.517 × 105 growth of Lactobacillus or Bifidobacteria in the midcolon (Chen et al.,
Da) and enzyme-extracted AXs (AXE) (MW = 3.252 × 104 Da). Com- 2015). Goblet cells synthesize and secret several mucins and protein
paring with AXA, AXE stimulated higher macrophage phagocytosis and barrier factors (Chen et al., 2015). The amount of mucin-degrading
showed a more delayed hypersensitivity reaction (Zhou et al., 2010). Akkermanis muciniphila decreases in cecal in AXs supplementation
Meanwhile, medium MW (1.56 × 105 Da) AXs isolated from barley feeding group (Van den Abbeele et al., 2011). Moreover, consumption
showed lower immunological responses (Samuelsen et al., 2011) while of the AXs-rich diet upregulates butyrate production leading to a bigger
arabinoxylan from the green leaves of Litsea glutinosa with a similar crypt depth, an indicator of enhancement of intestinal barrier function
level of MW (1.75 × 105 Da) showed stronger splenocyte, thymocyte, (Chen et al., 2015). Likewise, higher butyrate can upregulate the ex-
and macrophage activation (Das et al., 2013). pression of tight junction proteins, claudin 3 and elevate the production
MGN-3, a kind of AXs produced by enzymatic modification from of mucins, such as the dominant colonic mucin2 (Brahe, Astrup, &
rice bran, promotes the differentiation of human blood monocytes into Larsen, 2013; Neyrinck et al., 2011). Meanwhile, it is also possible that
immature dendritic cells through a reduction of CD14 monocyte ex- butyrate reduces intestinal permeability through a GLP-2 trophic effect
pression marker. Likewise, it triggers a significant increase in CD83, in the intestinal epithelium (Brahe et al., 2013). AXs modulate dis-
CD80, and the CD86 expression on immature dendritic cells and sti- ordered Cl- secretion in the mucosal epithelia to prevent excessive
mulated immature dendritic cell maturation, which in turn delivers co- water secretion, leading to a lower occurrence of pathophysiologic
stimulatory signals for effector T cells (Cholujova et al., 2013; Zhang, disease (Chen et al., 2015). AXs supplementation can also lower the
Li, et al., 2015). MGN-3 significantly promotes NK cell cytotoxicity, possibility of inflammation in the intestinal. It was reported that SCFAs,
activation, and expansion by inducing IFNγ, TNF-α, and CD54, CD69, especially butyrate, bound and stimulated nuclear transcription factor
CD25 receptors (Cholujova et al., 2013; Perez-Martinez et al., 2015). peroxisomeproliferator-activated receptor gamma, reduced the pro-in-
Furthermore, in the presence of IL-2, NK cytotoxicity can be increased flammatory nuclear transcription factor NFkβ by antagonizing its signal
(Cao et al., 2011; Perez-Martinez et al., 2015). Moreover, MGN-3 transduction (Nielsen, Theil, Purup, Norskov, & Bach Knudsen, 2015).
strengthens T and B cell functions, elevates lymphocyte ratio as well as AXOS supplementation also triggers a higher level of IL-10 in the serum
decreases the neutrophil ratio prominently (Kamiya et al., 2014; Zhang, to perform the anti-inflammatory effect.
Li, et al., 2015). Several studies investigated the anti-tumor property of Since AXs have been shown to modulate immune responses, it is
MGN-3. It increases the apoptosis of cancer cell lines by activating reasonable to speculate that they may function by acting like pathogen-
caspase 8, caspase 9, and caspase 3, depolarising mitochondrial mem- associated molecular (Zhang, Li, et al., 2015). This hypothesis was
brane potential, and inhibiting Bcl-2 (anti-apoptotic protein) expression confirmed by the notable immune-enhancing function of rice bran and
(Ghoneum & Gollapudi, 2003). Another alternative mechanism for corn husk derived AXs which resemble bacterial lipopolysaccharide in
MGN-3′s anti-tumor effects may be involving the activation of various terms of structure and MW (Zhang, Li, et al., 2015). This indicated that
endogenous antioxidant enzymes, for instance, GPx, SOD1, CAT, GST, the structure of AXs including chemical composition affected their
and reduction of lipid peroxidation (Zhang, Li, et al., 2015). Zheng, immune-enhancing function. Both AXs from banana peel hemicellulose
Sanada, Dohi, Hirai, and Egashira (2012) used 1N HCl to hydrolyzed and from corn hull contained galactose or glucuronic acid, which led to
MGN-3 at 100 °C for 1 h. The MGN-3 hydrolysate had the ability to strong immune responses (Ogawa et al., 2005; Zhang et al., 2004). It

544
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

avDP, average degree of polymerization; avDS, average degree of substitution; Ara/Xyl Ratio, arabinose/xylose ratio; MW, molecular weight; avDAS, average degree of arabinose substitution; AX, arabinoxylan; AXOS,
has been reported that the AXs contained galactose have the structural

(Malunga and Beta,


features that mainly affect CR3-expressing cells (Zhang, Li, et al., 2015).

(Ayala-Soto et al.,

(Ayala-Soto et al.,
(Bijalwan et al.,
(Malunga et al.,

(Snelders et al.,
(Yuwang et al.,
Comparing AXs from wheat with that from barley, the latter had a
lower MW and showed lower immunological responses in vitro, sug-
Reference

2018) gesting that MW may influenced the immune-enhancing function (Cao

2017)

2016)

2014)

2015)

2013)
2015)
et al., 2011; Samuelsen et al., 2011). It is reasonable to predict that
higher MW AXs have a greater chance to interact with different kinds of
receptors and their biological activity can last longer because higher
In vivo/in

In vitro

In vitro

In vitro

In vitro

In vitro

In vitro

In vitro
MW AXs requires a longer time to be degraded in mammalian systems
vitro

(Zhang, Li, et al., 2015). Other structural features, however, may offset
the effects of MW. Zhou et al. (2010) investigated the immune-enhan-
reducing ability; AXs with lower FA content exert higher toxicity against HepG2 than AXs

AXs with highest FA content shown highest antioxidant ability; AXs with low content of FA
but high content of uronic acid exhibited higher antioxidant ability than AX with medium
AXs with higher content of FA residues had higher antioxidant capacity; monosubstituted

AXs with highest total phenolic content possessed highest antioxidant ability followed by

cing function of AXA and AXE. The MW of AXA was 3.517 × 105 Da

Compared with free FA, bound FA residues demonstrate lower antioxidant capacity.The
AXs with highest branch exert lowest antioxidant effect; tannins contained AXs exhibit
AXs with higher FA content exhibited higher DPPH radical scavenging and ferric iron

without FA content, while the MW of AXE was 3.252 × 104 Da with

When DS and esterified FA content were relatively similar, the antioxidant capacity
declined with decreased DP.DS exerts negative effect on antioxidant capacity of AX

appearance of oxidative gelation lowers antioxidant capacity of feruloylated AXs.


high amount of FA (43.5 mg/100 g). The result turned out to be that
although AXA had higher MW than AXE, AXE stimulated a bigger
macrophage phagocytosis and delayed hypersensitivity reaction than
xylose residues relative proportions influenced the antioxidant capacity

AXA. It was possibly because the relatively higher amount of FA in AXE


allowed more cross-linkages between adjacent AXs chains thereby ex-
hibited a different tertiary conformation which might be easier to be
recognized by the receptors (Zhou et al., 2010). Similarly, Ma et al.
(2016) compared 3 types of water extractable AXs (WEAX) with dif-
ferent MW and FA content, they observed that the immunomodulatory
activity might be related to their content of FA more due to the anti-
AXs with second highest dehydrotri FA content.

oxidant activity and effect of prebiotic of FA. Moreover, AXs from


barley or from the green leaves of Litsea glutinosa had similar MW, but
the latter exerted a higher immunological response (Das et al., 2013;
Samuelsen et al., 2011). In AXs from barley, the arabinose residues
responsible for branching at backbone were either β- or α-anomer while
Effect on antioxidant capacity

highest antioxidant capacity.

in AXs from the green leaves of Litsea glutinosa, the arabinose residues,
responsible for branching at backbone were a mixture of β- and α-
with high FA content

anomers. This indicated that the difference of immune-enhancing


function between AXs from barley and from the green leaves of Litsea
glutinosa might due to different types of branching (Das et al., 2013). It
FA content.

has been widely known that MGN-3 has an extremely high immune-
enhancing function. The mechanisms underlying the im-
munomodulatory effect of MGN-3 is not clear yet, but one hypothesis is
that the structure of MGN-3 has some similarities with lipopoly-
AX1 (Ara/Xyl ratio, 0.53; highest total phenolic content and dehydrotri FA content) AX2
content of FA) AX3 (Ara/Xyl ratio, 1.09, lowest content of FA; higher content of uronic
AX1 (Ara/Xyl ratio, 0.85; high FA content) AX2 (Ara/Xyl ratio, 0.56; low FA content)
AX1 (Ara/Xyl ratio, 0.29; MW, 57 900 gmol−1; high FA content) AX2 (Ara/Xyl ratio,

AX1 (high bound FA content; avDP = 6.6; avDAS = 0.38) AX2 (high free FA content;
AX1 (Ara/Xyl ratio, 1.14; contained tannins) AX2 (Ara/Xyl ratio, 1.41) AX3 (Ara/Xyl
0.30; MW, 41,400 gmol−1; highest FA content) AX3 (Ara/Xyl ratio, 0.54; MW, 1520

(Ara/Xyl ratio, 0.5; second highest total phenolic content) AX3 (Ara/Xyl ratio, 0.55;

saccharide produced by gram-negative bacteria (Zhang, Li, et al.,


AX1 (Ara/Xyl ratio, 0.42; highest content of FA) AX2 (Ara/Xyl ratio, 0.59; medium

16 fractions of AXOS was collected with Ara/Xyl ratio ranged from 0.22 to 0.57

2015). Zheng et al. (2012) treated rats with MGN-3 or its low molecular
weighr hydrolysate (lower than 400 Da), then measured their hepato-
avDP = 6.6; avDAS = 0.39) AX3 (crosslinked; avDP = 3.8; avDAS = 0.23)

protective effect. The MGN-3 low molecular weighr hydrolysate had a


higher protective effect on D-GalN-induced hepatitis by reducing the IL-
lowest total phenolic content; second highest dehydrotriFA content)

8 concentration and its mRNA expression level, the effect compounds in


MGN-3 seemed to have low molecular weight. Zhang, Li, et al. (2015)
assumed that only when un-digestible AXs are degraded into smaller
molecules, can they easily pass through the intestinal wall and went
into the bloodstream. They are then transported to the lymph nodes to
Antioxidant capacity of AXs with different structural features.

exert the effect (Zhang, Li, et al., 2015).


In summary, the structural features of AXs relates to immune
function include MW, sugar composition, the existence of FA as well as
the type of branching. Higher molecule weight of AXs results in more
complicated tertiary structures of AXs, leading to an increase in the
arabinoxylooligosaccharides; FA, ferulic acid.

specific structural features which are able to interact with the receptors.
FA content will change the tertiary conformation through its cross-
gmol−1; low FA content)

linkage, which also contributes to immune modulation function. Above


Molecular structure

all, tertiary conformation characteristics play a significant role in AX’s


immune-enhancing function. Döring et al. (2016) showed that DS
ratio, 1.08)

would change the tertiary conformation of AXs, thus it can be hy-


pothesized that DS may be a contributor to modulate the immune re-
acid)

sponse. However, tertiary structure of AXs is not stable, it will change


because of intestinal environment. Likewise, body digesition and gut
Wheat aleurone

microbiota fermentation will break the tertiary structure. Thus, it is


Sorghum bran
Indian millet
Wheat bran

Wheat bran

worthwhile to figure out how much will the structure remain when AXs
Maize fiber
Rice bran
Sources

are degraded in intestine. FA, regarded as an important content in AXs,


Table 4

contributes to many physiological functions. Nevertheless, there are


only few studies focus on the effect of FA on the immune system.

545
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

Researches has been reported that phenolic acids such as flavonoids Barron, Gaiani, Dufour, & Micard, 2013).
were able to scavenge free radicals, thereby inhibit oxidative DNA Among the total phenolic acid of AXs, FA plays a significant role in
damage in lymphocytes, which may be one of the mechanism of the its antioxidant capacity. It has been proposed that FA content was the
effect of FA on the immune system (Park, Lee, Jeon, Jung, & Kang, major factor determining the antioxidant capacity of feruloylated AXOS
2010). (Malunga et al., 2017). Additionally, among FA, di-FA and tri-FA, all
esterified to arabinose, tri-FA had a higher antioxidant capacity since
5.4. Antioxidant capacity of AXs more units of FAs in each molecular elevated the hydrogen donor ca-
pacity of the moiety through providing more –OH groups (Ayala-Soto,
Recent studies reveal that the molecular structure, especially DS and Serna-Saldívar, García-Lara, & Pérez-Carrillo, 2014). As for the inter-
the MW, has a strong correlation with the antioxidant potential of AXs molecular reaction between feruloylated AXs, Snelders et al. (2013)
(Table 4) (Bijalwan, Ali, Kesarwani, Yadav, & Mazumder, 2016; observed a reduction of the antioxidant capacity and formation of
Malunga & Beta, 2015a; Malunga & Beta, 2015b; Yadav et al., 2017). oxidative gelation, probably attributed to dimerization of the FA moi-
As for DS, it has been reported that low branched hydroxycinnamic eties. Although oxidative gelation lowered antioxidant capacity of fer-
acid-bound arabinoxylans derived from kodo millet had a stronger uloylated AXs, it, however, enhanced the viscosity and improved some
antioxidant effect compared to hydroxycinnamic acid-bound arabi- other physiological functions (Aguedo et al., 2014).
noxylans derived from other sources which had a relatively higher DS Comparing with free FA, bound FA residues demonstrate a different
(Bijalwan et al., 2016). Likewise, a strong negative correlation between antioxidant capability due to the interaction between FA and AX moi-
DS and antioxidant capability was discovered in AXOS (Malunga & eties. FA which is esterified to AXs changes feruloylated AXs solubility,
Beta, 2015a). Both results suggested that the lower content of DS in which will influence the antioxidant effect of feruloylated AXs. Several
AXs, the stronger antioxidant capability. Researchers identify that the research groups reported that feruloylated AXs had a higher antioxidant
un-substituted xylose at O-2 or/and O-3 in AXs donated a hydrogen or capability when comparing to the similar amount of free FA. This result
an electron to participate in the antioxidant reaction (Malunga and was further confirmed by using ORAC and ABTS antioxidant assays
Beta, 2015a, 2016). Likewise, low DS increases the specific functional (performed in aqueous medium) (Malunga et al., 2017). The possible
groups of xylan with metal-binding activity (for example, –COOH and mechanism is that the hydrophily of FA is increased when FA is ester-
–OH) which are the most efficient inhibitors of hydroxyl radical, be- ified to arabinose, which increases the effective concentration of FA in a
cause metal ions can be the catalysts in hydroxyl radical production water medium. Similarly, in physiological conditions (mostly in aqu-
(Rivas, Conde, Moure, Dominguez, & Parajo, 2013). Nevertheless, eous medium), feruloylated AXs have higher antioxidant capacity
Malunga et al. (2017) found out that WEAX from wheat with higher DS (Malunga et al., 2017). When the assays performed in oil-water
had a higher antioxidant activity. The WEAX fractions with higher DS medium, the antioxidant effects of feruloylated AXs in the oxidation
was easier to disperse into the reaction mixture because the steric system of LDL and lipid were higher compared to free FA (Ayala-Soto
hindrance caused by the presence of mono- or di-substituted xylose et al., 2014; Snelders et al., 2014). It is because esterified FA provided a
prevented intermolecular cross-linking, leading to better participation lipophilic part to hydrophilic AXs, facilitated their interaction with
of AXs in the redox reaction systems (Malunga et al., 2017). oxidant factors located in the interface or in the oil phase. In vivo ex-
Several experiments have been done to investigate the effects of the periments demonstrated that during interaction with microbiota, the
MW of AXs on their antioxidant properties. Rao and Muralikrishna FA esterase activity in the inoculum was hindered by the presence of
(2006) reported that feruloylated AXs with higher MW also had a bound FA through steric hindrance, thus, less FA was released during
higher antioxidant capacity. It was also observed that when the content incubation (Snelders et al., 2014). Free FA was further metabolized into
of DS was similar to the content of esterified FA in AXs, the antioxidant less antioxidative 4-vinylguaiacol and 3-phenylpropionic (Snelders
capacity declined with decreased DP (Malunga & Beta, 2015a). Ele- et al., 2014). Therefore, in the colon, bound FA can prevent it from
vated (1–4)-β-D-xylopyranose linkages (in the case of increased DP) being destructed by gut microbiota because of its molecular and special
lead to a higher amount of unsubstituted xylose, which can participate microstructure, results in a sustained antioxidant capacity (Snelders
in antioxidant reaction through donating a hydrogen or an electron and et al., 2014). In contrast, Rivas et al. (2013) found that compared with
thereby increase the potency of AXs and AXOS to against free radicals. free FA, the ability of ferulated AXs to protect β-carotene-linoleic
In contrast, Malunga et al. (2017) treated the WEAX fractions with emulsions from oxidation was lower. It is because feruloylated oligo-
endoxylanase to reduce their MWs. The results indicated that partial saccharides are more hydrophilic compared to free FA, which will
depolymerization of WEAX doubled their antioxidant activity, which hinder their interaction with the oil-water interface in the emulsion,
could be attributed to their higher mobility in the solution and conse- thus reduces the emulsifying capacity. Lower emulsifying capacity ne-
quently enhanced the radical-scavenging activity (Malunga et al., gatively affects the partition coefficient between the aqueous and the
2017). Additionally, the pattern of arabinose substitution and the ex- lipophilic phase, which is reported to affect its antioxidant capacity
istence of acetyl and uronyl groups may also influence the antioxidant (Snelders et al., 2013). Similarly, the viability of HepG2 cells and their
activity of AXs (Malunga et al., 2017; Rivas et al., 2013). IC50 after treatment with AXs fractions showed that feruloylated AXs
To make a conclusion, researches shows that the effect of DS and containing mostly free FA exhibited higher hydrophobicity, and might
MW on AXs antioxidant ability are inconsistent. It is because that DS be deeply embedded in membranes (oil medium). Therefore they could
and the MW of AXs not only influenced the specific functional groups of break oxidative chain reaction and exhibited high antioxidant capacity
AXs but also impacted the interaction between AXs molecules through (Oteiza, Erlejman, Verstraeten, Keen, & Fraga, 2005; Yuwang et al.,
steric hindrance as well as between AXs and the solvent. It should take 2018).
all into consideration when examing the specifical effects of DS and the It can be concluded that the feruloylated AXs demonstrates incon-
MW on the antioxidant capacity of AXs. sistent antioxidant capacity in water, oil-water and oil medium re-
The antioxidant capacity correlates with the total phenolic content spectively. The different solubility of feruloylated AXs in each medium
of each AX fraction (Bijalwan et al., 2016; Malunga & Beta, 2015b; can be one of the reasons. Thus, to exam the antioxidant capacity of
Yuwang et al., 2018). The phenolic acid of AXs mostly is covalently feruloylated AXs under physiological condition, the solubility of fer-
bounded to arabinose residue, generally including a large amount of uloylated AXs in different systems is of great importance to investigate.
FA, some p-coumaric acid, and a little caffeic acid. Some special kinds Moreover, AXs with high free FA content leading to high solubility in
of AXs also have phytic acid, tannin, and anthocyanins (Aguedo, oil medium should be put into consideration, for it may be deeply
Fougnies, Dermience, & Richel, 2014; Ayala-Soto et al., 2015; Bijalwan embedded in the membrane to effectively perform an antioxidant
et al., 2016; Malunga et al., 2016; Revanappa & Salimath, 2011; Rosa, ability. The molecular and tertiary conformation of feruloylated AXs

546
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

will hinder the structural transformation during gut microbiota fer- functional group exert a remarkable impact on the antioxidant capacity,
mentation and consequently improve the antioxidant availability in the which correlates with DS, MW and content of total phenolic acid
colon. In the colon, FA in bound or free form, without being degraded especially FA. The relationship between antioxidant capacity and each
by gut microbiota, can directly exert high antioxidant ability on colon structural feature should not be analyzed separately, but rather should
epithelium cells, thereby may be able to decrease colorectal cancer all be taken into consideration. In addition, because AX’s antioxidant
(Snelders et al., 2014). At the same time, the structure of saccharide ability would be greatly influenced by the surrounding medium, the
moiety of feruloylated AXs was observed to change the bioavailability antioxidant availability in physiological conditions is of great im-
in plasma. When rats fed with free FA, the concentration of FA present portance. Overall, among all the structural features of AXs, molecule
in plasma in the free and conjugated forms was higher but for a shorter weight, DS and FA content remarkably influence their physiological
time compared with rats fed with bound FA (Zhaohui, Yukari, & Hiroo, functions in a complex way.
2003). Thus, when to determine the antioxidant capacity of fer- FA is of great significance in the physiological functions of AXs
uloylated AXs, we should take not simply its FA content into con- especially in AXs’ antioxidant capacity. Likewise, it exhibits a cross-
sideration, but also the solubility, MW as well as special tertiary con- linking effect to highly improve the viscosity of AXs, which is able to
formation. Since the bound FA content and free FA content of modulate lipid and glucose metabolism. However, up to now, the in-
feruloylated AXs influence antioxidant capacity differently, further formation is not enough to compare the contribution of FA content and
studies should be performed to study the balance between bound FA AX moiety in feruloylated AXs on physiological functions. There are
and free FA content to obtain specific feruloylated AXs with highest only few studies concentrate on the ability of FA to change composition
antioxidant capacity in physiological condition. and abundance of gut microbiota. The contribution of FA on modula-
Generally, the p-coumaric acid content in AXs is the second highest tion of immune system is still not clear. Furthermore, it has been re-
phenolic acid followed by FA in AXs (Yuwang et al., 2018). Ayala-Soto ported FA would distribute to bloodstream or to peripheral tissue after
et al. (2014) even showed that p-coumaric exerted a higher antioxidant absorption, and it is possible that the FA distributed to different part of
capacity compared to the same amount of FA. High tannin sorghum the body results in different health benefits. At the same time, the
brans containing high tannin and nine 3-deoxyanthocyanins are known structure of saccharide moiety of feruloylated AXs was observed to
to have higher antioxidant properties than other phenols and are cur- change the bioavailability in plasma as well as in colon which may
rently being considered as effective nutraceuticals against oxidative change the physiological functions of AXs. Further studies may need to
stress (Ayala-Soto et al., 2015). AXs from destarched wheat bran con- find out the structure of saccharide which in favor of bioavailability of
tain a high amount of phytic acid. Although phytic acid is a chelator of FA.
several minerals of nutritional importance, resulting in a decrease in
minerals bioavailability and dietary deficiencies, it can be a source of
phosphate and is thought to be anti-carcinogenic thanks to its anti- 7. Conclusion
oxidant properties (Aguedo et al., 2014). Further research shall be able
to find out the exact contribution of phytic acid in AXs to human health. Above all, the interaction between AXs and gut microbiota changes
Above all, both the content of total phenolic acid and AXs structure SCFAs production as well as the composition and abundance of colonic
contribute to the antioxidant capacity of feruloylated AXs. Compared microbiota, both impact human health. At the same time, AXs de-
the two factors, the antioxidant capacity of feruloylated AXs was mainly monstrated various physiological functions. However, the specific in-
dependent on FA content, regardless of bound FA or free FA. However, fluence on each physiological functions need to be improve by further
the role of AXs moiety of feruloylated AXs in antioxidant capacity researches. As for the effects on lipid and glucose metabolism, there are
shouldn’t be underestimated. We hypothesized that the hydroxyl groups lack of studies to investigate the effects of AXs themselves on the me-
of unsubstituded xylose AXs moiety may participate in antioxidant re- tabolism, including the mechanism of AXs to change the gene expres-
action, and AXs moiety may the influence the antioxidant ability of FA. sion. Likewise, the antioxidant capacity under inner environment of
Both of them may be the mechnisms of AXs moiety ability to change the human body is still unclear. When it comes to the direct effect of AXs on
antioxidant ability of feruloylated. So far, there are plenty of studies physiological functions, it is important to find out the bioactive fraction
focus on the antioxidant ability of AXs, however, most studies con- of AXs. However, there are few studies focus on this fraction. It has been
ducted in vitro and the antioxidant effects of feruloylated AXs in vivo reported the immunoregulation effect compounds in MGN-3 seemed to
are still unclear. Further studies may need to focus on the degradation have low molecular weight (lower than 400 Da). Thus, it may be hy-
of feruloylated AXs in intestine and determine the antioxidant effect in pothesised that the bioactive fractions which have the ability to mod-
vivo, since digestion and interaction with gut microbiota exert large ulate the gene expression in liver or absorped into plasma may have the
effects on feruloylated AXs antioxidant capacity. similar molecular weight as the effect compounds in MGN-3. AX’s
structural characteristics have a big impact on those physiological
6. The relationship between structure and physiological functions functions and interaction with gut microbiota. Molecule weight, DS,
and FA content are three main factors influence their physiological
Different AXs structure exert different physiological function functions. The structure characteristics can be one of the factors to
(Fig. 3). It can be concluded that AXs with lower molecule weight, analyze physiological functions of different kinds of AXs from various
lower DS, less FA content and simple linkage, and sugar pattern lead to sources or extracted by diverse methods. Future researches can design
a higher fermentation rate and SCFA production. Comparing with AXs, particular method to obtain AXs with specific structure in order to ac-
AXOS tends to stimulate the growth of bifidobacteria. When it comes to quire products for health-improving or for food application.
lipids metabolism and glucose metabolism, the viscosity related struc-
tures are more important which including MW, arabinose: xylose ratio, 8. Declaration
the degree of branching and the content of bound FA. AXs with higher
MW, the degree of branching and bound FA content will cause higher The author declares no conflict of interest.
viscosity. However, AXs with lower or higher viscosity both have some
benefits to human. As for immune-enhancing function, AXs with higher
MW, higher content of FA or inclusion of galactose or glucuronic acid 9. Ethics statements
can strongly enhance immune function. The spatial arrangement of AXs
plays an important role in the immune-enhancing function. Moreover, This research did not include any human subjects and animal ex-
emulsifying capacity, solubility, and the amount of antioxidant periments.

547
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

Viscosity
Solubility
Immune-enhancing ability Lipid and glucose metabolism Viscosity
The amount of SCFAs The amount of SCFA The amount of SCFA
Antioxidant capacity Lipid and glucose metabolism
Antioxidant capacity
substitution Immune-enhancing ability

Glca D-Gal α-L-Ara α-L-Ara


Xyl - Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl – Xyl –Xyl – Xyl
β-L-Ara - α-L-Ara - α-L-Ara α-L-Ara Gal – Ara
Type of O Substitution
branching pattern

The amount of SCFAs The amount of SCFA


Immune-enhancing ability Antioxidant capacity

Viscosity
Solubility
The amount of SCFA
Lipid and glucose metabolism
Antioxidant capability
Immune-enhancing ability
Fig. 3. Overall representation of different impacts on physiological functions of each AX structural features. MW, FA content and substitution (including ara/xyl
ratio, degree of substitution) are the three main factors affecting physiological functions, exerting effects on all physiological functions and physicochemical
properties mentioned in this article. Special sugar composition such as galactose and glucuronic impacts AX immune-enhancing ability and the amount of produced
SCFAs, similar as the effects of type of branching. At the same time, AX substitution pattern (including different substitution site) change the amount of produced
SCFAs and its antioxidant capacity.

Acknowledgment byproducts have prebiotic potential in mice fed a high-fat diet. Journal of Agriculture
and Food Chemistry, 62, 8169–8178.
Bijalwan, V., Ali, U., Kesarwani, A. K., Yadav, K., & Mazumder, K. (2016).
This work was supported by Sichuan Science and Technology Hydroxycinnamic acid bound arabinoxylans from millet brans-structural features and
Program (Grant No. 2018JY0472), the State Key Program of National antioxidant activity. International Journal of Biological Macromolecules, 88, 296–305.
Natural Science of China (Grant No. C170105) and Scientific Research Biliaderis, C. G., & Izydorczyk, M. S. (2007). Functional food carbohydrates. FL: CRC Press.
Boll, E. V., Ekstrom, L. M., Courtin, C. M., Delcour, J. A., Nilsson, A. C., Bjorck, I. M., et al.
Training Program for Undergraduates of Sichuan Agricultural (2016). Effects of wheat bran extract rich in arabinoxylan oligosaccharides and re-
University (Grant No. 2019529). sistant starch on overnight glucose tolerance and markers of gut fermentation in
healthy young adults. European Journal of Nutrition, 55, 1661–1670.
Brahe, L. K., Astrup, A., & Larsen, L. H. (2013). Is butyrate the link between diet, in-
References testinal microbiota and obesity-related metabolic diseases? Obesity Reviews, 14,
950–959.
Aguedo, M., Fougnies, C., Dermience, M., & Richel, A. (2014). Extraction by three pro- Broekaert, W. F., Courtin, C. M., Verbeke, K., Van de Wiele, T., Verstraete, W., & Delcour,
cesses of arabinoxylans from wheat bran and characterization of the fractions ob- J. A. (2011). Prebiotic and other health-related effects of cereal-derived arabinox-
tained. Carbohydrate Polymers, 105, 317–324. ylans, arabinoxylan-oligosaccharides, and xylooligosaccharides. Critical Reviews in
Akhtar, M., Tariq, A. F., Awais, M. M., Iqbal, Z., Muhammad, F., Shahid, M., et al. (2012). Food Science and Nutrition, 51, 178–194.
Studies on wheat bran Arabinoxylan for its immunostimulatory and protective effects Buksa, K., Nowotna, A., Praznik, W., & Gambuś, H. (2012). Isolation, modification and
against avian coccidiosis. Carbohydrate Polymers, 90, 333–339. characterization of soluble arabinoxylan fractions from rye grain. European Food
Andrewartha, K. A., Phillips, D. R., & Stone, B. A. (1979). Solution properties of wheat- Research and Technology, 235, 385–395.
flour arabinoxylans and enzymically modified arabinoxylans. Carbohydrate Research, Cao, L., Liu, X., Qian, T., Sun, G., Guo, Y., Chang, F., et al. (2011). Antitumor and im-
77, 191–204. munomodulatory activity of arabinoxylans: A major constituent of wheat bran.
Audrey, R., Marija, S., David, L., Frédéric, L., & Luc, D. V. (2016). Bifidobacteria and International Journal of Biological Macromolecules, 48, 160–164.
butyrate-producing colon bacteria: Importance and strategies for their stimulation in Chassard, C., Goumy, V., Leclerc, M., Del'Homme, C., & Bernalier-Donadille, A. (2010).
the human Gut. Frontiers in Microbiology, 7. Characterization of the xylan-degrading microbial community from human faeces.
Ayala-Soto, F. E., Serna-Saldívar, S. O., García-Lara, S., & Pérez-Carrillo, E. (2014). FEMS Microbiology Ecology, 61, 121–131.
Hydroxycinnamic acids, sugar composition and antioxidant capacity of arabinoxylans Chen, H., Fu, Y., Jiang, X., Li, D., Qin, W., Zhang, Q., et al. (2018). Arabinoxylan activates
extracted from different maize fiber sources. Food Hydrocolloids, 35, 471–475. lipid catabolism and alleviates liver damage in rats induced by high-fat diet. Journal
Ayala-Soto, F. E., Serna-Saldívar, S. O., Welti-Chanes, J., & Gutierrez-Uribe, J. A. (2015). of the Science of Food and Agriculture, 98, 253–260.
Phenolic compounds, antioxidant capacity and gelling properties of glucoarabinox- Chen, H., Wang, W., Degroote, J., Possemiers, S., Chen, D., De Smet, S., et al. (2015).
ylans from three types of sorghum brans. Journal of Cereal Science, 65, 277–284. Arabinoxylan in wheat is more responsible than cellulose for promoting intestinal
Banuls, C., Rovira-Llopis, S., Lopez-Domenech, S., Veses, S., Victor, V. M., Rocha, M., barrier function in weaned male piglets. Journal of Nutrition, 145, 51–58.
et al. (2016). Effect of consumption of a carob pod inositol-enriched beverage on Chen, T., Long, W., Zhang, C., Shuang, L., Zhao, L., & Hamaker, B. R. (2017). Fiber-
insulin sensitivity and inflammation in middle-aged prediabetic subjects. Food & utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota.
Function, 7, 4379–4387. Scientific Reports-UK, 7, 2594.
Bauman, D. E., Harvatine, K. J., & Lock, A. L. (2011). Nutrigenomics, rumen-derived Cherrington, C. A., Hinton, M., Pearson, G. R., & Chopra, I. (2010). Short-chain organic
bioactive fatty acids, and the regulation of milk fat synthesis. Annual Review of acids at ph 5.0 kill Escherichia coli and Salmonella spp. without causing membrane
Nutrition, 31, 299–319. perturbation. Journal of Applied Bacteriology, 70, 161–165.
Berger, K., Falck, P., Linninge, C., Nilsson, U., Axling, U., Grey, C., et al. (2014). Cereal Cholujova, D., Jakubikova, J., Czako, B., Martisova, M., Hunakova, L., Duraj, J., et al.

548
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

(2013). MGN-3 arabinoxylan rice bran modulates innate immunity in multiple carbohydrates including arabinoxylans of different structure. Journal of the Science of
myeloma patients. Cancer Immunology, Immunotherapy, 62, 437–445. Food and Agriculture, 80, 1211–1218.
Christensen, K. L., Hedemann, M. S., Laerke, H. N., Jorgensen, H., Mutt, S. J., Herzig, K. Grootaert, C., Abbeele, P. V. D., Marzorati, M., Broekaert, W. F., Courtin, C. M., Delcour,
H., et al. (2013). Concentrated arabinoxylan but not concentrated beta-glucan in J. A., et al. (2009). Comparison of prebiotic effects of arabinoxylan oligosaccharides
wheat bread has similar effects on postprandial insulin as whole-grain rye in porto- and inulin in a simulator of the human intestinal microbial ecosystem. FEMS
arterial catheterized pigs. Journal of Agriculture and Food Chemistry, 61, 7760–7768. Microbiology Ecology, 69, 231–242.
Cja, V., Delcour, J. A., Verbruggen, M. A., & Gruppen, H. (1995). Rye water-soluble Gunness, P., Flanagan, B. M., Mata, J. P., Gilbert, E. P., & Gidley, M. J. (2016). Molecular
arabinoxylans also vary in their 2-monosubstituted xylose content. Cereal Chemistry, interactions of a model bile salt and porcine bile with (1,3:1,4)-beta-glucans and
72, 227–228. arabinoxylans probed by (13)C NMR and SAXS. Food Chemistry, 197, 676–685.
Courtin, C. M. (2011). Prebiotic and other health-related effects of cereal-derived arabi- Gunness, P., Flanagan, B. M., Shelat, K., Gilbert, R. G., & Gidley, M. J. (2012). Kinetic
noxylans, arabinoxylan-oligosaccharides, and xylooligosaccharides. Critical Reviews analysis of bile salt passage across a dialysis membrane in the presence of cereal
in Food Science and Nutrition, 51, 178–194. soluble dietary fibre polymers. Food Chemistry, 134, 2007–2013.
Courtin, C. M., & Delcour, J. A. (2002). Arabinoxylans and endoxylanases in wheat flour Gunness, P., Williams, B. A., Gerrits, W. J., Bird, A. R., Kravchuk, O., & Gidley, M. J.
bread-making. Journal of Cereal Science, 35, 225–243. (2016). Circulating triglycerides and bile acids are reduced by a soluble wheat ara-
Damen, B., Verspreet, J., Pollet, A., Broekaert, W. F., Delcour, J. A., & Courtin, C. M. binoxylan via modulation of bile concentration and lipid digestion rates in a pig
(2011). Prebiotic effects and intestinal fermentation of cereal arabinoxylans and model. Molecular Nutrition & Food Research, 60, 642–651.
arabinoxylan oligosaccharides in rats depend strongly on their structural properties Hartvigsen, M. L., Gregersen, S., Laerke, H. N., Holst, J. J., Bach Knudsen, K. E., &
and joint presence. Molecular Nutrition & Food Research, 55, 1862–1874. Hermansen, K. (2014). Effects of concentrated arabinoxylan and beta-glucan com-
Das, D., Maiti, S., Maiti, T. K., & Islam, S. S. (2013). A new arabinoxylan from green leaves pared with refined wheat and whole grain rye on glucose and appetite in subjects
of Litsea glutinosa (Lauraeae): Structural and biological studies. Carbohydrate with the metabolic syndrome: A randomized study. European Journal of Clinical
Polymers, 92, 1243–1248. Nutrition, 68, 84–90.
Dervilly-Pinel, G., Tran, V., & Saulnier, L. (2004). Investigation of the distribution of Hartvigsen, M. L., Jeppesen, P. B., Laerke, H. N., Njabe, E. N., Knudsen, K. E., &
arabinose residues on the xylan backbone of water-soluble arabinoxylans from wheat Hermansen, K. (2013). Concentrated arabinoxylan in wheat bread has beneficial
flour. Carbohydrate Polymers, 55, 171–177. effects as rye breads on glucose and changes in gene expressions in insulin-sensitive
Dervillypinel, G., Rimsten, L., Saulnier, L., Andersson, R., & Aman, P. (2001). Water- tissues of Zucker diabetic fatty (ZDF) rats. Journal of Agriculture and Food Chemistry,
extractable arabinoxylan from pearled flours of wheat, barley, rye and triticale. 61, 5054–5063.
Evidence for the presence of ferulic acid dimers and their involvement in gel for- Heimann, E., Nyman, M., & Degerman, E. (2015). Propionic acid and butyric acid inhibit
mation. Journal of Cereal Science, 34, 207–214. lipolysis and de novo lipogenesis and increase insulin-stimulated glucose uptake in
Desai, M. S., Seekatz, A. M., Koropatkin, N. M., Kamada, N., Hickey, C. A., Wolter, M., primary rat adipocytes. Adipocyte, 4, 81–88.
et al. (2016). A dietary fiber-deprived gut microbiota degrades the colonic mucus Henderson, A. J., Kumar, A., Barnett, B., Dow, S. W., & Ryan, E. P. (2012). Consumption
barrier and enhances pathogen susceptibility. Cell, 167, 1339–1353. of rice bran increases mucosal immunoglobulin A concentrations and numbers of
Dhital, S., Dolan, G., Stokes, J. R., & Gidley, M. J. (2014). Enzymatic hydrolysis of starch intestinal Lactobacillus spp. Journal of Medicinal Food, 15, 469–475.
in the presence of cereal soluble fibre polysaccharides. Food & Function, 5, 579–586. Hermes, R. G., Manzanilla, E. G., Martin-Orue, S. M., Perez, J. F., & Klasing, K. C. (2011).
Dodd, D., Mackie, R. I., & Cann, I. K. O. (2011). Xylan degradation, a metabolic property Influence of dietary ingredients on in vitro inflammatory response of intestinal por-
shared by rumen and human colonic Bacteroidetes. Molecular Microbiology, 79, cine epithelial cells challenged by an enterotoxigenic Escherichia coli (K88).
292–304. Comparative Immunology, Microbiology and Infectious Diseases, 34, 479–488.
Döring, C., Jekle, M., & Becker, T. (2016). Technological and analytical methods for Hopkins, M. J., Englyst, H. N., Macfarlane, S., Furrie, E., Macfarlane, G. T., & Mcbain, A.
Arabinoxylan quantification from cereals. Critical Reviews in Food Science and J. (2003). Degradation of cross-linked and non-cross-linked arabinoxylans by the
Nutrition, 56, 999–1011. intestinal microbiota in children. Applied and Environmental Microbiology, 69,
Druart, C., Neyrinck, A. M., Dewulf, E. M., De Backer, F. C., Possemiers, S., Van de Wiele, 6354–6360.
T., et al. (2013). Implication of fermentable carbohydrates targeting the gut micro- Hosoda, Y., Okahara, F., Mori, T., Deguchi, J., Ota, N., Osaki, N., et al. (2017). Dietary
biota on conjugated linoleic acid production in high-fat-fed mice. British Journal of steamed wheat bran increases postprandial fat oxidation in association with a re-
Nutrition, 110, 998–1011. duced blood glucose-dependent insulinotropic polypeptide response in mice. Food &
Ebringerová, A., Hromádková, Z., Petráková, E., & Hricovíni, M. (1990). Structural fea- Nutrition Research, 61, 1361778.
tures of a water-soluble L-arabino-D-xylan from rye bran. Carbohydrate Research, Hosseini, E., Grootaert, C., Verstraete, W., & Van de Wiele, T. (2011). Propionate as a
198, 57. health-promoting microbial metabolite in the human gut. Nutrition Reviews, 69,
Eeckhaut, V., Van, I. F., Dewulf, J., Pasmans, F., Haesebrouck, F., Ducatelle, R., et al. 245–258.
(2008). Arabinoxylooligosaccharides from wheat bran inhibit Salmonella coloniza- Ingerslev, A. K., Theil, P. K., Hedemann, M. S., Lærke, H. N., & Bach Knudsen, K. E.
tion in broiler chickens. Poultry Science, 87, 2329. (2014). Resistant starch and arabinoxylan augment SCFA absorption, but affect
Fadel, A., Mahmoud, A. M., Ashworth, J. J., Li, W., Ng, Y. L., & Plunkett, A. (2017). postprandial glucose and insulin responses differently. British Journal of Nutrition,
Health-related effects and improving extractability of cereal arabinoxylans. 111, 1564–1576.
International Journal of Biological Macromolecules. Izydorczyk, M. S., & Biliaderis, C. G. (1992). Effect of molecular size on physical prop-
Fry, S. C. (2004). Primary cell wall metabolism: Tracking the careers of wall polymers in erties of wheat arabinoxylan. Journal of Agriculture and Food Chemistry, 40, 561–568.
living plant cells. New Phytologist, 161, 641–675. Izydorczyk, M. S., & Biliaderis, C. G. (1993). Cereal arabinoxylans: Advances in structure
Fu, J., Wei, B., Wen, T., Johansson, M. E., Liu, X., Bradford, E., et al. (2011). Loss of and physicochemical properties. Carbohydrate Polymers, 28, 33–48.
intestinal core 1-derived O-glycans causes spontaneous colitis in mice. Journal of Izydorczyk, M. S., & Biliaderis, C. G. (1995). Cereal arabinoxylans: Advances in structure
Clinical Investigation, 121, 1657. and physicochemical properties. Carbohydrate Polymers, 28, 33–48.
Garcia, A. L., Otto, B., Reich, S. C., Weickert, M. O., Steiniger, J., Machowetz, A., et al. Izydorczyk, M. S., & Dexter, J. E. (2008). Barley β-glucans and arabinoxylans: Molecular
(2007). Arabinoxylan consumption decreases postprandial serum glucose, serum structure, physicochemical properties, and uses in food products – A review. Food
insulin and plasma total ghrelin response in subjects with impaired glucose tolerance. Research International, 41, 850–868.
European Journal of Clinical Nutrition, 61, 334–341. Izydorczyk, M. S., & Macgregor, A. W. (2000). Evidence of intermolecular interactions of
Garcia, A. L., Steiniger, J., Reich, S. C., Weickert, M. O., Harsch, I., Machowetz, A., et al. beta-glucans and arabinoxylans. Carbohydrate Polymers, 41, 417–420.
(2006). Arabinoxylan fibre consumption improved glucose metabolism, but did not Jiang, Y., Jo, A. Y., & Graff, J. M. (2012). SnapShot: Adipocyte life cycle. Cell, 150
affect serum adipokines in subjects with impaired glucose tolerance. Hormone and 234–234 e232.
Metabolic Research, 38, 761–766. Jurgoński, A., Krotkiewski, M., Juśkiewicz, J., & Billing-Marczak, K. (2015). Suppression
Gemen, R., de Vries, J. F., & Slavin, J. L. (2011). Relationship between molecular of postprandial glycaemia by L-arabinose in rats is more associated with starch than
structure of cereal dietary fiber and health effects: Focus on glucose/insulin response sucrose ingestion – Short report. Polish Journal of Food and Nutrition Sciences, 65.
and gut health. Nutrition Reviews, 69, 22–33. Juvonen, K. R., Purhonen, A. K., Salmenkallio-Marttila, M., Lahteenmaki, L., Laaksonen,
Geraylou, Z., Souffreau, C., Rurangwa, E., Maes, G. E., Spanier, K. I., Courtin, C. M., et al. D. E., Herzig, K. H., et al. (2009). Viscosity of oat bran-enriched beverages influences
(2013). Prebiotic effects of arabinoxylan oligosaccharides on juvenile Siberian stur- gastrointestinal hormonal responses in healthy humans. Journal of Nutrition, 139,
geon (Acipenser baerii) with emphasis on the modulation of the gut microbiota using 461–466.
454 pyrosequencing. FEMS Microbiology Ecology, 86, 357–371. Kamiya, T., Shikano, M., Tanaka, M., Ozeki, K., Ebi, M., Katano, T., et al. (2014).
Geurts, L., Neyrinck, A. M., Delzenne, N. M., Knauf, C., & Cani, P. D. (2014). Gut mi- Therapeutic effects of biobran, modified arabinoxylan rice bran, in improving
crobiota controls adipose tissue expansion, gut barrier and glucose metabolism: symptoms of diarrhea predominant or mixed type irritable bowel syndrome: A pilot,
Novel insights into molecular targets and interventions using prebiotics. Beneficial randomized controlled study. Evidence-Based Complementary and Alternative Medicine,
Microbes, 5, 3–17. 2014, 828137.
Ghoneum, M., & Gollapudi, S. (2003). Modified arabinoxylan rice bran (MGN-3/Biobran) Kieffer, D. A., Piccolo, B. D., Marco, M. L., Kim, E. B., Goodson, M. L., Keenan, M. J., et al.
sensitizes human T cell leukemia cells to death receptor (CD95)-induced apoptosis. (2016). Obese mice fed a diet supplemented with enzyme-treated wheat bran display
Cancer Letters, 201, 41–49. marked shifts in the liver metabolome concurrent with altered gut bacteria. Journal of
Giulia Falchi, A., Grecchi, I., Muggia, C., Palladini, G., & Perlini, S. (2016). Effects of a Nutrition, 146, 2445–2460.
bioavailable Arabinoxylan-enriched white bread flour on postprandial glucose re- Kimura, I., Ozawa, K., Inoue, D., Imamura, T., Kimura, K., Maeda, T., et al. (2013). The
sponse in normoglycemic subjects. Journal of Dietary Supplements, 13, 626–633. gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty
Glenwright, A. J., Pothula, K. R., Bhamidimarri, S. P., Chorev, D. S., Baslé, A., Firbank, S. acid receptor GPR43. Nature Communications, 4, 1829.
J., et al. (2017). Structural basis for nutrient acquisition by dominant members of the Köhnke, T., Ostlund, A., & Brelid, H. (2011). Adsorption of arabinoxylan on cellulosic
human gut microbiota. Nature, 541, 407. surfaces: Influence of degree of substitution and substitution pattern on adsorption
Glitso, L. V., Jensen, B. B., & Bach Knudsen, K. E. (2000). In vitro fermentation of rye characteristics. Biomacromolecules, 12, 2633.

549
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

Koropatkin, N. M., Cameron, E. A., & Martens, E. C. (2012). How glycan metabolism Park, Y. K., Lee, H. B., Jeon, E. J., Jung, H. S., & Kang, M. H. (2010). Chaga mushroom
shapes the human gut microbiota. Nature Reviews Microbiology, 10, 323–335. extract inhibits oxidative DNA damage in human lymphocytes as assessed by comet
Lambertusam, V. D. B., & Alphonsgj, V. (2008). Bifidobacterium glycoside hydrolases and assay. BioFactors, 21, 109–112.
(potential) prebiotics. Innovative Food Science and Emerging, 9, 401–407. Perez-Martinez, A., Valentin, J., Fernandez, L., Hernandez-Jimenez, E., Lopez-Collazo, E.,
Lopez, H. W., Levrat, M. A., Guy, C., Messager, A., Demigné, C., & Rémésy, C. (1999). Zerbes, P., et al. (2015). Arabinoxylan rice bran (MGN-3/Biobran) enhances natural
Effects of soluble corn bran arabinoxylans on cecal digestion, lipid metabolism, and killer cell-mediated cytotoxicity against neuroblastoma in vitro and in vivo.
mineral balance (Ca, Mg) in rats. Journal of Nutritional Biochemistry, 10, 500. Cytotherapy, 17, 601–612.
Lu, Z. X., Walker, K. Z., Muir, J. G., & O'Dea, K. (2004). Arabinoxylan fibre improves Pollet, A., Craeyveld, V. V., Wiele, T. V. D., Verstraete, W., Delcour, J. A., & Courtin, C. M.
metabolic control in people with Type II diabetes. European Journal of Clinical (2012). In vitro fermentation of arabinoxylan oligosaccharides and low molecular
Nutrition, 58, 621. mass arabinoxylans with different structural properties from wheat (Triticum aes-
Ma, X., Wang, L., Wei, H., Huo, X., Wang, C., Liu, D., et al. (2016). Adjuvant properties of tivum L.) bran and psyllium (Plantago ovata Forsk) seed husk. Journal of Agriculture
water extractable arabinoxylans with different structural features from wheat flour and Food Chemistry, 60, 946–954.
against model antigen ovalbumin. Food & Function, 7, 1537–1543. Qiang, X., Chao, Y. L., & Wan, Q. B. (2009). Health benefit application of functional
Malunga, L. N., & Beta, T. (2015a). Antioxidant capacity of arabinoxylan oligosaccharide oligosaccharides. Carbohydrate Polymers, 77, 435–441.
fractions prepared from wheat aleurone using Trichoderma viride or Neocallimastix Rao, R. S., & Muralikrishna, G. (2006). Water soluble feruloyl arabinoxylans from rice and
patriciarum xylanase. Food Chemistry, 167, 311–319. ragi: Changes upon malting and their consequence on antioxidant activity.
Malunga, L. N., & Beta, T. (2015b). Antioxidant capacity of water-extractable arabinox- Phytochemistry, 67, 91–99.
ylan from commercial barley, wheat, and wheat fractions. Cereal Chemistry, 92 Revanappa, S. B., & Salimath, P. V. (2011). Phenolic acid profiles and antioxidant ac-
141002053404000. tivities of different wheat (Triticum Aestivum L.) varieties. Journal of Food
Malunga, L. N., & Beta, T. (2016). Isolation and identification of feruloylated arabinox- Biochemistry, 35, 759–775.
ylan mono- and oligosaccharides from undigested and digested maize and wheat. Rivas, S., Conde, E., Moure, A., Dominguez, H., & Parajo, J. C. (2013). Characterization,
Heliyon, 2, e00106. refining and antioxidant activity of saccharides derived from hemicelluloses of wood
Malunga, L. N., Eck, P., & Beta, T. (2016). Inhibition of intestinal alpha-glucosidase and and rice husks. Food Chemistry, 141, 495–502.
glucose absorption by feruloylated arabinoxylan mono- and oligosaccharides from Rivière, A., Moens, F., Selak, M., Maes, D., Weckx, S., & De, V. L. (2014). The ability of
corn bran and wheat aleurone. Journal of Nutrition and Metabolism, 2016, 1932532. bifidobacteria to degrade arabinoxylan oligosaccharide constituents and derived
Malunga, L. N., Izydorczyk, M., & Beta, T. (2017). Effect of water-extractable arabinox- oligosaccharides is strain dependent. Applied and Environmental Microbiology, 80,
ylans from wheat aleurone and bran on lipid peroxidation and factors influencing 204–217.
their antioxidant capacity. Bioactive Carbohydrates and Dietary Fibre, 10, 20–26. Rosa, N. N., Barron, C., Gaiani, C., Dufour, C., & Micard, V. (2013). Ultra-fine grinding
Martens, E. C., Koropatkin, N. M., Smith, T. J., & Gordon, J. I. (2011). Complex glycan increases the antioxidant capacity of wheat bran. Journal of Cereal Science, 57, 84–90.
catabolism by the human gut microbiota: The Bacteroidetes Sus-like paradigm. Rose, D. J., Patterson, J. A., & Hamaker, B. R. (2010). Structural differences among alkali-
Journal of Biological Chemistry, 284, 24673. soluble arabinoxylans from maize (Zea mays), rice (Oryza sativa), and wheat
Mccleary, B.V., Prosky, L. (2008). Advanced Dietary Fibre Technology. (Triticum aestivum) brans influence human fecal fermentation profiles. Journal of
Mendis, M., Leclerc, E., & Simsek, S. (2016). Arabinoxylans, gut microbiota and im- Agricultural and Food Chemistry, 58, 493–499.
munity. Carbohydrate Polymers, 139, 159–166. Rumpagaporn, P., Reuhs, B. L., Kaur, A., Patterson, J. A., Keshavarzian, A., & Hamaker, B.
Mendis, M., Martens, E. C., & Simsek, S. S. (2018). How fine structural differences of xylo- R. (2015). Structural features of soluble cereal arabinoxylan fibers associated with a
oligosaccharides and arabinoxylo-oligosaccharides regulate differential growth of slow rate of in vitro fermentation by human fecal microbiota. Carbohydrate Polymers,
Bacteroides species. Journal of Agriculture and Food Chemistry acs.jafc.8b01263-. 130, 191.
Michlmayr, H., Hell, J., Lorenz, C., Böhmdorfer, S., Rosenau, T., & Kneifel, W. (2013). Samuelsen, A. B., Rieder, A., Grimmer, S., Michaelsen, T. E., & Knutsen, S. H. (2011).
Arabinoxylan oligosaccharide hydrolysis by family 43 and 51 glycosidases from Immunomodulatory activity of dietary fiber: Arabinoxylan and mixed-linked beta-
Lactobacillus brevis DSM 20054. Applied and Environmental Microbiology, 79, glucan isolated from barley show modest activities in vitro. International Journal of
6747–6754. Molecular Sciences, 12, 570–587.
Möhlig, M., Koebnick, C., Weickert, M. O., Lueder, W., Otto, B., Steiniger, J., et al. (2005). Sanchez, J. I., Marzorati, M., Grootaert, C., Baran, M., Craeyveld, V. V., Courtin, C. M.,
Arabinoxylan-enriched meal increases serum ghrelin levels in healthy humans. et al. (2009). Arabinoxylan-oligosaccharides (AXOS) affect the protein/carbohydrate
Hormone and Metabolic Research, 37, 303. fermentation balance and microbial population dynamics of the Simulator of Human
Ndeh, D., & Gilbert, H. J. (2018). Biochemistry of complex glycan depolymerisation by Intestinal. Microbial Ecology, 2, 101–113.
the human gut microbiota. FEMS Microbiology Review, 42, 2. Sarma, S. M., Singh, D. P., Singh, P., Khare, P., Mangal, P., Singh, S., et al. (2018). Finger
Neyrinck, A. M., Hiel, S., Bouzin, C., Campayo, V. G., Cani, P. D., Bindels, L. B., et al. millet arabinoxylan protects mice from high-fat diet induced lipid derangements,
(2018). Wheat-derived arabinoxylan oligosaccharides with bifidogenic properties inflammation, endotoxemia and gut bacterial dysbiosis. International Journal of
abolishes metabolic disorders induced by western diet in mice. Nutrition & Diabetes, Biological Macromolecules, 106, 994–1003.
8, 15. Shelat, K. J., Vilaplana, F., Nicholson, T. M., Wong, K. H., Gidley, M. J., & Gilbert, R. G.
Neyrinck, A. M., Possemiers, S., Druart, C., Van de Wiele, T., De Backer, F., Cani, P. D., (2010). Diffusion and viscosity in arabinoxylan solutions: Implications for nutrition.
et al. (2011). Prebiotic effects of wheat arabinoxylan related to the increase in bifi- Carbohydrate Polymers, 82, 46–53.
dobacteria, Roseburia and Bacteroides/Prevotella in diet-induced obese mice. PLoS Sheridan, P. O., Martin, J. C., Lawley, T. D., Browne, H. P., Harris, H. M.,
ONE, 6, e20944. Bernalierdonadille, A., et al. (2016). Polysaccharide utilization loci and nutritional
Neyrinck, A. M., Van Hee, V. F., Piront, N., De Backer, F., Toussaint, O., Cani, P. D., et al. specialization in a dominant group of butyrate-producing human colonic Firmicutes.
(2012). Wheat-derived arabinoxylan oligosaccharides with prebiotic effect increase Microbial Genomics, 2.
satietogenic gut peptides and reduce metabolic endotoxemia in diet-induced obese Shiyi Ou, G. M. J., Jiao, X., Chen, J., Jianzhong Wu, A., & Huang, X. S. (2007). Protection
mice. Nutrition & Diabetes, 2, e28. against oxidative stress in diabetic rats by wheat bran feruloyl oligosaccharides.
Nie, Q., Chen, H., Hu, J., Gao, H., Fan, L., Long, Z., & Nie, S. (2018). Arabinoxylan at- Journal of Agricultural and Food Chemistry, 55, 3191.
tenuates type 2 diabetes by improvement of carbohydrate, lipid, and amino acid Snelders, J., Dornez, E., Delcour, J. A., & Courtin, C. M. (2013). Ferulic Acid content and
metabolism. Molecular Nutrition & Food Research, 1800222. appearance determine the antioxidant capacity of arabinoxylanoligosaccharides.
Nielsen, K. L., Hedemann, M. S., Laerke, H. N., Jorgensen, H., & Bach Knudsen, K. E. Journal of Agricultural and Food Chemistry, 61, 10173–10182.
(2014). A metabolomics approach used to profile plasma from portal-arterial pigs Snelders, J., Olaerts, H., Dornez, E., Van de Wiele, T., Aura, A.-M., Vanhaecke, L., et al.
revealed differences between breads incurred by dietary fibre and protein contents. (2014). Structural features and feruloylation modulate the fermentability and evo-
Journal of Nutritional Science, 3, e18. lution of antioxidant properties of arabinoxylanoligosaccharides during in vitro fer-
Nielsen, T. S., Theil, P. K., Purup, S., Norskov, N. P., & Bach Knudsen, K. E. (2015). Effects mentation by human gut derived microbiota. Journal of Functional Foods, 10, 1–12.
of resistant starch and arabinoxylan on parameters related to large intestinal and Suriano, F., Bindels, L. B., Verspreet, J., Courtin, C. M., Verbeke, K., Cani, P. D., et al.
metabolic health in pigs fed fat-rich diets. Journal of Agriculture and Food Chemistry, (2017). Fat binding capacity and modulation of the gut microbiota both determine
63, 10418–10430. the effect of wheat bran fractions on adiposity. Scientific Reports, 7, 5621.
Niewold, T. A., Schroyen, M., Geens, M. M., Verhelst, R. S. B., & Courtin, C. M. (2012). Tong, L. T., Zhong, K., Liu, L., Qiu, J., Guo, L., Zhou, X., et al. (2014). Effects of dietary
Dietary inclusion of arabinoxylan oligosaccharides (AXOS) down regulates mucosal wheat bran arabinoxylans on cholesterol metabolism of hypercholesterolemic ham-
responses to a bacterial challenge in a piglet model. Journal of Functional Foods, 4, sters. Carbohydrate Polymers, 112, 1–5.
626–635. Van den Abbeele, P., Gerard, P., Rabot, S., Bruneau, A., El Aidy, S., Derrien, M., et al.
Nyman, E. M. (2002). Importance of processing for physico-chemical and physiological (2011). Arabinoxylans and inulin differentially modulate the mucosal and luminal
properties of dietary fibre. Proceedings of the Nutrition Society, 62, 187–192. gut microbiota and mucin-degradation in humanized rats. Environmental
Ogawa, K., Takeuchi, M., & Nakamura, N. (2005). Immunological effects of partially Microbiology, 13, 2667–2680.
hydrolyzed arabinoxylan from corn husk in mice. Bioscience, Biotechnology, and Vangsøe, C. T., Ingerslev, A. K., Theil, P. K., Hedemann, M. S., Lærke, H. N., & Knudsen, K.
Biochemistry, 69, 19–25. E. B. (2016). In vitro starch digestion kinetics of diets varying in resistant starch and
Oteiza, P. I., Erlejman, A. G., Verstraeten, S. V., Keen, C. L., & Fraga, C. G. (2005). arabinoxylan compared with in vivo portal appearance of glucose in pigs. Food
Flavonoid-membrane interactions: A protective role of flavonoids at the membrane Research International, 88, 199–206.
surface? Clinical and Developmental Immunology, 12, 19–25. Vanhamel, S., Cleemput, G., Delcour, J. A., Nys, M., & Darius, P. L. (1993).
Pantophlet, A. J., Wopereis, S., Eelderink, C., Vonk, R. J., Stroeve, J. H., Bijlsma, S., et al. Physicochemical and functional properties of rye nonstarch polysaccharides. IV. The
(2017). Metabolic profiling reveals differences in plasma concentrations of arabinose effect of high MW water-soluble pentosans on wheat-bread quality in a straight-
and xylose after consumption of fiber-rich pasta and wheat bread with differential dough procedure. Cereal Chemistry, 70, 306–311.
rates of systemic appearance of exogenous glucose in healthy men. Journal of Vardakou, M., Palop, C. N., Christakopoulos, P., Faulds, C. B., Gasson, M. A., & Narbad, A.
Nutrition, 147, 152–160. (2008). Evaluation of the prebiotic properties of wheat arabinoxylan fractions and

550
Z. Chen et al. Journal of Functional Foods 54 (2019) 536–551

induction of hydrolase activity in gut microflora. International Journal of Food Zhang, D., Williams, B. A., Mikkelsen, D., Li, X., Keates, H. L., Lisle, A. T., et al. (2015).
Microbiology, 123, 166–170. Soluble arabinoxylan alters digesta flow and protein digestion of red meat-containing
Vogel, B., Gallaher, D. D., & Bunzel, M. (2012). Influence of cross-linked arabinoxylans on diets in pigs. Nutrition, 31, 1141–1147.
the postprandial blood glucose response in rats. Journal of Agriculture and Food Zhang, P., Wampler, J. L., Bhunia, A. K., Burkholder, K. M., Patterson, J. A., & Whistler, R.
Chemistry, 60, 3847–3852. L. (2004). Effects of arabinoxylans on activation of murine macrophages and growth
Wood, P. J., Braaten, J. T., Scott, F. W., Riedel, K. D., Wolynetz, M. S., & Collins, M. W. performance of broiler chicks. Cereal Chemistry, 81, 511–514.
(1994). Effect of dose and modification of viscous properties of oat gum on plasma Zhang, S., Li, W., Smith, C. J., & Musa, H. (2015). Cereal-derived arabinoxylans as bio-
glucose and insulin following an oral glucose load. British Journal of Nutrition, 72, logical response modifiers: Extraction, molecular features, and immune-stimulating
731–743. properties. Critical Reviews in Food Science and Nutrition, 55, 1035–1052.
Yadav, M. P., Kale, M. S., Hicks, K. B., & Hanah, K. (2017). Isolation, characterization and Zhaohui, Z., Yukari, E., & Hiroo, S. (2003). Ferulic acid sugar esters are recovered in rat
the functional properties of cellulosic arabinoxylan fiber isolated from agricultural plasma and urine mainly as the sulfoglucuronide of ferulic acid. Journal of Nutrition,
processing by-products, agricultural residues and energy crops. Food Hydrocolloid, 63, 133, 1355.
545–551. Zheng, S., Sanada, H., Dohi, H., Hirai, S., & Egashira, Y. (2012). Suppressive effect of
Yuwang, P., Sulaeva, I., Hell, J., Henniges, U., Bohmdorfer, S., Rosenau, T., et al. (2018). modified arabinoxylan from rice bran (MGN-3) on D-galactosamine-induced IL-18
Phenolic compounds and antioxidant properties of arabinoxylan hydrolysates from expression and hepatitis in rats. Bioscience, Biotechnology, and Biochemistry, 76,
defatted rice bran. Journal of the Science of Food and Agriculture, 98, 140–146. 942–946.
Zeng, H., Xue, Y., Peng, T., & Shao, W. (2007). Properties of xylanolytic enzyme system in Zhou, S., Liu, X., Guo, Y., Wang, Q., Peng, D., & Cao, L. (2010). Comparison of the im-
bifidobacteria and their effects on the utilization of xylooligosaccharides. Food munological activities of arabinoxylans from wheat bran with alkali and xylanase-
Chemistry, 101, 1172–1177. aided extraction. Carbohydrate Polymers, 81, 784–789.

551

View publication stats

You might also like