You are on page 1of 20

Critical Reviews in Food Science and Nutrition

ISSN: 1040-8398 (Print) 1549-7852 (Online) Journal homepage: http://www.tandfonline.com/loi/bfsn20

Intestinal in vitro cell culture models and their


potential to study the effect of food components
on intestinal inflammation

Maria del Carmen Ponce de León-Rodríguez, Jean-Pierre Guyot & Caroline


Laurent-Babot

To cite this article: Maria del Carmen Ponce de León-Rodríguez, Jean-Pierre Guyot & Caroline
Laurent-Babot (2018): Intestinal in�vitro cell culture models and their potential to study the effect of
food components on intestinal inflammation, Critical Reviews in Food Science and Nutrition, DOI:
10.1080/10408398.2018.1506734

To link to this article: https://doi.org/10.1080/10408398.2018.1506734

Published online: 02 Oct 2018.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=bfsn20
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION
https://doi.org/10.1080/10408398.2018.1506734

REVIEW

Intestinal in vitro cell culture models and their potential to study


the effect of food components on intestinal inflammation
n-Rodrıguez, Jean-Pierre Guyot, and Caroline Laurent-Babot
Maria del Carmen Ponce de Leo
NUTRIPASS—University of Montpellier, IRD, Montpellier SupAgro, Montpellier, France

ABSTRACT KEYWORDS
Cell cultures are widely used in pharmaceutical, medical, food/nutrition and biological sciences. In Bioactive compounds; gut;
food and nutrition science, intestinal cell culture models of human origin are attracting increasing intestinal epithelial cells;
interest but are still rarely used in investigations of the effects of bioactive food compounds on immune cells
intestinal inflammation. However, such in vitro models would, among other benefits, limit the use
of in vivo models and could provide new molecular data.
This review is an overview of two-dimensional (2D) and three-dimensional (3D) intestinal cell cul-
ture models and their potential use in gut inflammation studies. After describing the features of
healthy and inflamed intestinal barriers, we describe the main intestinal cell lines (Caco2, HT29,
T84) and their use in investigations of the transport and antioxidant/anti-inflammatory potential of
some bioactive food compounds. Finally, different co-culture models of gut inflammation, in asso-
ciation with immune cells (PBMC, THP1 and RAW 264.7 cell lines) in both 2D and 3D models are
presented. 3D models called organs-on-chips or biochips are the most recent and very promising
approach made possible by bioengineering and biotechnological improvements and more accur-
ately mimic the gut microenvironment.

Introduction components, including bioactive compounds with potential


risks or benefits for health. Among these compounds, phyto-
The intestinal epithelium is a permeable barrier that plays a
chemicals such as polyphenols or carotenoids have been
major role in the regulation of solute and fluid exchange
shown to have anti-inflammatory and antioxidant effects
and has direct consequences for the absorption and trans-
and benefits for chronic disorders.
port of nutrients. Different types of cells (enterocytes, goblet In recent decades, in vitro, ex vivo and in vivo models
cells, M cells, Paneth cells and others such as endocrine and have been developed to study different intestinal epithelium
immune cells) form the intestinal epithelium. Beneath the functions and metabolism, in particular in the inflamed
epithelial monolayer, immune cells (e.g. macrophages, lym- state. Although animal models can simulate the physiology
phocytes) are spread into the lamina propria. Good cooper- of an entire organism, the main disadvantages of in vivo
ation between all these cells maintains mucosal integrity, models are the variation in responses due to differences
which is challenged daily by external factors in the luminal between species and the difficulty in extrapolating results to
environment. The loss of the intestinal barrier function con- humans (Fitzgerald et al. 2015). In addition, the use of cell
sequently has clinical and nutritional consequences including culture in vitro models has increased because the use of ani-
chronic disorders, inflammation and malnutrition. The eti- mal models involves ethical issues and is quite expensive.
ology of gut inflammation, whose incidence and prevalence Cell culture models have been widely used in bioavailability
is increasing worldwide, is still not fully understood, and toxicological studies in both the food and pharmaceut-
although multiple causes are known to be involved ical fields. Improved in vitro models using cells of a human
(Molodecky et al. 2012; Loftus 2004). Low or high grade gut origin have the advantage of reducing animal experimenta-
inflammation is often associated with other pathological tion and could enable the study of molecular mechanisms in
conditions (e.g. infectious diseases, inflammatory bowel dis- a simple and reproducible way. For instance, the molecular
eases) but also with metabolic syndromes and with some study of bioactive food compounds and their interactions
noncommunicable diseases (NCDs) (e.g. obesity) (Ding and with the intestinal epithelial barrier and microbiota has
Lund 2011; Esser et al. 2014; Walters, Xu, and Knight 2014). become possible.
Moreover, malnutrition is a common feature related to the Originally, cell lines were used in cancer research but
inflammatory status of the intestine (Benjamin et al. 2008; later some of these models were further used for in-depth
Forbes et al. 2017; Massironi et al. 2013; Mijac et al. 2010). investigations of physiological mechanisms thanks to their
The intestinal epithelium is intensely exposed to food ability to differentiate into cells expressing normal

CONTACT Caroline Laurent-Babot caroline.laurent@umontpellier.fr NUTRIPASS—University of Montpellier, IRD, Montpellier SupAgro, Montpellier, France
ß 2018 Taylor & Francis Group, LLC
2 
M. C. PONCE DE LEON-RODRIGUEZ ET AL.

characteristics. In addition, co-culture models have been Schulzke 2009). Hence, food molecules and diverse microor-
developed to better mimic the complex composition of the ganisms, including microbiota promote sustained activation
intestinal barrier as described above. Most currently of the mucosal immune system (Geremia et al. 2014;
employed cell culture models are performed in two-dimen- Shanahan and Quigley 2014; Vindigni et al. 2016).
sional (2D) conditions with one or more cell lines in (co- The epithelial intercellular junctions that form the apical
)culture, and frequently use insert systems composed of rigid junction complex (AJC) are crucial to preserve the intestinal
porous membranes. The intrinsic limitations of those models barrier function. The AJC formed by transmembrane proteins
drove the development of three-dimensional (3D) culture is represented by the tight junctions (TJs, e.g. occludin and
systems. As a consequence of technological improvements, claudins) and the adherens junctions (AJs, e.g. E-cadherin)
the use of 3D in vitro models to reconstitute the in vivo cel- that either interact with the cytoskeleton via cytoplasmic pro-
lular microenvironment has increased significantly. teins like the zonula occludens proteins (ZO) or the catenins
In the context of the extensive use of cell cultures in family. Paracellular transport is regulated by the TJs, which
many different fields, this review provides an overview of seals the space between adjacent epithelial cells and control
2D and 3D intestinal cell culture models and their potential the passage of molecules based on size and charge selectivity.
use in gut inflammation studies. These models have great Moreover, the active nutrient transport pathway (e.g., Naþ-
potential for studies of the effect of bioactive food com- co-transporters), depends on the transepithelial concentration
pounds on intestinal inflammation. gradient generated by the flux of ions through the TJs (Liang
and Weber 2014; Turner 2009). Alterations in permeability
will result in loss of water in addition to malabsorption of
Features of healthy and inflamed intestinal barrier
nutrients, therefore, homeostasis depends to a great extent on
Characterized by a single-cell layer, the intestinal epithelium a thoroughly functional TJ system.
is mainly composed of polarized absorptive cells called In addition to behaving as a physical barrier, the intestinal
enterocytes. However, other specialized epithelial cells are epithelium also plays a major role in the host defense system.
also present including goblet cells, microfold cells (M cells) As described above, immune cells are present in the lamina
and Paneth cells, forming the whole gut wall. Goblet cells propria (Peyer’s patches), making the gut the main immune
are responsible for the production of mucins that form a organ of the human body. In healthy circumstances, the intes-
protective mucus layer and Paneth cells secrete antimicrobial tinal barrier is in a necessary and continuous state of
peptides (defensins) and growth factors. The intestinal epi- “balanced” inflammation with a fragile equilibrium that can
thelium is an important barrier between the lumen and the be altered by several factors. Homeostasis can be disrupted by
intestinal lamina propria and functions as a dynamic inter- both acute and chronic processes. For example, during intes-
face between the external environment and intestinal tissues tinal infections, pathogens such as Salmonella spp,
(Viladomiu et al. 2013; Wehkamp et al. 2005). These cells Escherichia coli, Clostridium difficile or Giardia often destabil-
all cooperate to prevent the entrance of microorganisms and ize the host TJ system and subsequently cause episodes of
other particles into the host and are also involved in epithe- diarrhea (Nusrat et al. 2001; O’Hara and Buret 2008;
lial growth and repair maintaining the mucosal integrity. Simonovic et al. 2000). The etiology of intestinal chronic dis-
Beneath the intestinal epithelial monolayer, immune cells orders such as IBD is however, not yet clear, although it is
(macrophages, dendritic cells, lymphocytes) are spread into certain that multiple causes are involved. Ulcerative colitis
the lamina propria. These defense cells are more concen- (UC) and Crohn’s disease (CD) are the main inflammatory
trated in Peyer’s patches (secondary lymphoid organ) and bowel diseases studied, as they have the highest prevalence
interact with epithelial cells via soluble factors like growth worldwide (Loftus Jr 2004; Molodecky et al. 2012).
factors, cytokines and enzymes (Abraham and Cho 2009). Many studies suggest that alterations in TJs and subse-
The intestinal epithelium is a permeable barrier that also quent changes in permeability in IBD exacerbate the inflam-
plays a major role in the regulation of solute and fluid matory process as the result of an imbalance between pro-
exchange with direct consequences for the absorption and inflammatory and anti-inflammatory cytokines produced by
transport of nutrients. This necessary exchange occurs via immune cells (Neurath 2014; Pandolfi et al. 2009; Suzuki
intercellular junctions present between the cells, which allow 2013). It has been demonstrated that cytokines like TNF-a,
paracellular transport. Accordingly, direct cell damage INF-c, IL-1b and IL-13 induce intestinal epithelial barrier
observed for example in inflammatory bowel diseases (IBD), dysfunction which increases the paracellular infiltration of
provokes loss of barrier function. Clinical evidence has shown luminal potentially antigenic molecules (Al-Sadi et al. 2011;
that malnutrition is a common feature in IBD patients Heller et al. 2005; Turner 2006; Utech et al. 2005; Wang
(Benjamin et al. 2008; Forbes et al. 2017; Massironi et al. et al. 2005; Watson et al. 2005). This suggests that the intes-
2013; Mijac et al. 2010). Diverse micronutrient deficiencies tinal barrier function plays a crucial role in IBD pathophysi-
e.g. minerals (Fe, Zn), vitamins (D, A, B9, B12) and proteins ology, but it remains unclear whether these changes are part
have been reported in patients with chronic intestinal disor- of the cause or the consequence of chronic inflammation.
ders (Conklin and Oliva-Hemker 2010; Mallon and Suskind Likewise, different studies have shown the effects of diet on
2010; Vagianos et al. 2007). Additionally, changes in perme- TJ integrity (Kosi nska and Andlauer 2013; Suzuki 2013).
ability can cause closer interaction between immune cells of According to in vitro studies, dietary components such as
the lamina propria and the lumen content (Hering and polyphenols or casein peptides may improve the intestinal
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 3

barrier function, whereas others like ethanol and acetalde- nutrition since the establishment of in vitro cell lines from
hyde its metabolite, are thought to have a negative impact both animal and human origin. The use of primary cell cul-
(Ferguson 2014; Samak, Aggarwal, and Rao 2011; Shimizu tures is limited by their availability, repeatability and their
2017; Suzuki, Tanabe, and Hara 2011; Yasumatsu and use in long term studies because of their short life span, but
Tanabe 2010). transformed cell lines are largely used instead (Langerholc
Recent reports strongly suggest that intestinal microbiota et al. 2011; Zucco et al. 2005). The development of in vitro
play an important role in preserving gut health. A disturb- models to mimic the in vivo environment required the char-
ance in the equilibrium of microbiota composition may rep- acterization of cell lines as single or co-culture models
resent a risk of infections developing (Khosravi and before they could be used successfully. In this context,
Mazmanian 2013). This imbalance can also modify the human cell lines have been favored.
response of the intestinal immune system and the epithelial
barrier function that subserve the development of IBD and
could even have extraintestinal consequences (Brown, Main intestinal cell lines
Sadarangani, and Finlay 2013; Cammarota et al. 2015; Caco-2 cell line
Fyderek et al. 2009; Kamada et al. 2013; Kostic, Xavier, and The human epithelial cell line Caco-2 has been the cell line
Gevers 2014; Manichanh et al. 2012; Reiff and Kelly 2010). most widely used as an intestinal model for absorption,
Changes in microbiota composition were also linked to transport and bioavailability studies in recent decades
reduced short-chain-fatty acids (SCFAs) metabolism which are (Cheng, Li, and Uss 2008; Hidalgo, Raub, and Borchardt
known to have anti-inflammatory and immune-regulatory 1989; Hillgren, Kato, and Borchardt 1995). This cell line was
activity (Arpaia et al. 2013; Maslowski et al. 2009; Smith et al. isolated from a human colorectal adenocarcinoma and
2013; Vieira et al. 2012). Despite limited available information, established by Fogh & Trempe in 1974 (Fogh and Trempe
there are reports concerning dietary impact on intestinal 1975). It is well characterized as an enterocyte model thanks
microbiota and the link that those changes might have with to its morphological and functional features expressed after
IBD (Sheehan, Moran, and Shanahan 2015; Wu et al. 2011; differentiation. It takes these cells about five days to reach
Zimmer et al. 2012). As mentioned above, the intestinal epithe- confluence and spontaneously start to differentiate during
lium plays an important role in immunity. Macrophages and 30 days of culture. Once differentiated, they form a polarized
dendritic cells present in the lamina propria are responsible for cell monolayer with apical and basolateral membranes, a
sensing luminal antigens while regulatory T lymphocytes junction complex and a brush border with microvilli on the
(Treg) maintain a balance by suppressing abnormal immune apical side typical of human enterocytes (Chantret et al.
response against the microbiota or dietary antigens (Geremia 1988; Hidalgo, Raub, and Borchardt 1989; Pinto et al. 1983).
et al. 2014; Strober, Fuss, and Blumberg 2002). It is thus clear The expression of enzymes such as hydrolases (sucrose-iso-
that altered interactions between intestinal microbiota and the maltase, lactase, etc.) present in intestinal microvilli
immune response also play an important role in intestinal (Chantret et al. 1994; Howell, Kenny, and Turner 1992;
inflammation (Kamada and N ~ez 2014). However, further
un Vachon and Beaulieu 1992) and different membrane trans-
studies are needed to clearly understand this relationship. porters are also traits of the Caco-2 cell line (Lea 2015).
Based on epidemiological studies, changes in food con- However, the Caco-2 parental cell line has certain limita-
sumption with increased intake of animal proteins, n-6 fatty tions including the formation of a heterogeneous monolayer
acids and ‘fast food’ among other dietary components, are linked to culture time and to the number of passages
thought to be linked to changes in microbiota and to the (Artursson, Palm, and Luthman 2001) and the presence of
risk of developing IBD. In contrast, a Mediterranean diet, multilayer zones (Briske-Anderson, Finley, and Newman
rich in vegetables, fruit, cereals and fish, but also in olive oil 1997). The cultivation conditions may also select subpopula-
and red wine has been reported to reduce the risk of IBD. tions of cells (Lea 2015). In addition, this cell line cannot
However, no causal food item has been identified in studies differentiate into goblet cells, so one important limitation is
of the association between diet and IBD due to interpret- their inability to produce mucus.
ation difficulties (Ng et al. 2013). In addition to dietary Diverse Caco-2 clones have been established to reduce
factors, TJ and immune dysregulation and a potential micro- heterogeneity (e.g. Caco-2/TC7, Caco-2/15, Caco-2/AQ)
biota disequilibrium, genetic, environmental and socioeco- (Sambuy et al. 2005). The TC7 clone was derived from a
nomic factors may also influence the development of IBD late passage (198) of the Caco-2 line and differentiates faster
and are reviewed elsewhere (Cho 2008; Khor, Gardet, and than the parental line (20 vs. 30 days) by retaining its mor-
Xavier 2011; Liu and Stappenbeck 2016; Ng et al. 2013). phological and functional characteristics and the monolayer
formed is more homogeneous and stable (Caro et al. 1995;
I. Chantret et al. 1994; Ranaldi et al. 2003; Turco et al.
In vitro cell culture models of gut inflammation
2011). Moreover, the TC7 clone has been shown to be a
Technological advances in cell culture in the last 40 years suitable model of the small intestinal barrier in terms of
have increased research into intestinal disorders and physio- reproducibility and performance (Zucco et al. 2005).
logical epithelial barrier mechanisms with the aim of mini- Caco-2 cells have also been shown to be able to produce
mizing in vivo experiments prior to clinical trials. Cell inflammatory markers (e. g., cytokines) in response to other
culture has become an essential tool in toxicology and cytokines such as IL-1b, TNF-a and/or IFNc (Hollebeeck
4 
M. C. PONCE DE LEON-RODRIGUEZ ET AL.

et al. 2012; Rodrıguez-Ramiro et al. 2013; WeR glarz et al. goblet-like mucus-producing cells (Lesuffleur et al. 1990),
2007) and/or in some cases, in response to external stimuli which have been employed as a model to study intestinal
like lipopolysaccharides (LPS) that are present on the mem- cell adhesion of lactic acid bacteria (Turpin et al. 2012).
branes of microorganisms (Barrera and Sanchez 2015; The HT29-MTX has also been widely used in co-culture
Maccaferri et al. 2012; WeR glarz et al. 2007; Xu et al. 2012). systems along with Caco-2 cell line to mimic the human
It should be pointed out that reports also showed Caco-2 intestinal barrier in absorption and permeability studies
cells were hypo-responsive to LPS stimulation due to the (Barnett et al. 2016; Beduneau et al. 2014; Behrens et al.
limited expression of the Toll-like receptor involved in LPS 2001; Kaulmann, Andre, et al. 2016; Laparra, Glahn, and
recognition (TLR-4), even though these cells express the co- Miller 2009; Mahler, Shuler, and Glahn 2009; Nollevaux
receptor MD-2, which is essential for NF-jB activation and et al. 2006; Poquet, Clifford, and Williamson 2008; Selby-
the subsequent secretion of pro-inflammatory cytokines Pham et al. 2017). Other mucus secreting cell lines have
(Suzuki, Hisamatsu, and Podolsky 2003; Vamadevan been characterized including LS174T and LS180 (Tom et al.
et al. 2010). 1976) and clones from these lines have been isolated to
study mucins (Kuan et al. 1987).
HT-29 cell line
The HT-29 is another intestinal cell line, established by T84 cell line
Fogh and Trempe. This cell line was also derived from a A third cell line frequently used in intestinal studies is T84,
colon adenocarcinoma (Fogh and Trempe 1975) and has which was established in 1980 by Murakami et Masui to
been used as an enterocyte model in bioavailability and cell study the hormonal control of human colon carcinoma cell
mechanism studies (Andoh et al. 2001; Hagesaether 2011; Yi growth (Murakami and Masui 1980). T84 cells grow in
et al. 2016). The HT-29 line is considered as a pluripotent monolayers of polarized cells that show the presence of TJs
intestinal cell line because changes in the culture media can and small microvilli on the surface (Dharmsathaphorn et al.
lead to different enterocytic differentiation paths. Unlike in 1984). Unlike spontaneous differentiation of Caco-2, a par-
the Caco-2 cell line, in HT-29, differentiation is not spon- ticularity of the T84 cell line is that differentiation into
taneous but rather depends on nutritional and culture con- crypt-like cells (Madara and Dharmsathaphorn 1985) com-
ditions (Huet et al. 1987; Viallard et al. 1986; Zweibaum mences only after induction with human recombinant trans-
et al. 2011). In a glucose free medium, in a similar way to forming growth factor (TGFb1) or other soluble factors
the Caco-2 line, HT-29 cells differentiate into enterocyte- from mesenchymal cells. This feature makes it possible to
type cells expressing hydrolases like sucrose-isomaltase, study the differentiation process (Juuti-Uusitalo et al. 2006).
which are restricted to the intestinal brush border, whereas T84 cells have also been used to study electrolyte trans-
when grown in presence of glucose, they remain undifferen- port mechanisms (Alzamora et al. 2011; Beltran et al. 2015;
tiated. Furthermore, the differentiation in HT-29 cells can Nichols et al. 2015; Tang et al. 2010) and intestinal perme-
be inhibited and reversed by the addition of glucose to the ability (Ewaschuk et al. 2008; Huang et al. 2016; Watson
medium (Zweibaum et al. 1985). Other differences between et al. 2005). Besides, this cell line has also been employed in
Caco-2 and HT-29 lines are a longer differentiation period studies of signaling pathways related to the inflammatory
and the absence of lactase expression in the HT-29 line. response of intestinal epithelium (Cario et al. 2000; Cho and
Nevertheless, the main difference between HT-29 and Caco- Park 2015; de Kivit et al. 2011; Juuti-Uusitalo et al. 2011;
2 cell lines is that under certain culture conditions, HT-29 Ou et al. 2009; Vamadevan et al. 2010) and bacteria-entero-
can differentiate into goblet-like cells and thus has the abil- cyte interaction (Otte and Podolsky 2004).
ity to produce mucus (Zweibaum et al. 2011). The mucus Despite the wide use of Caco-2, HT29 and T84, other
produced by goblet cells is a water insoluble gel mainly human gut cell lines (e.g. SW480, H4) and animal cell mod-
composed of glycoprotein oligomers and related monomers els are available; some of them are reviewed elsewhere
that forms the protective layer in the intestine (Beduneau (Langerholc et al. 2011; Leibovitz et al. 1976). A gene
et al. 2014). expression profile of some of these commonly exploited
When treated with sodium-butyrate, for example, HT-29 intestinal cell lines was published in 2012 to help choose the
cells differentiate into distinct phenotypes, as demonstrated appropriate in vitro model for specific studies (Bourgine
by Augeron and Laboisse (1984), who isolated different et al. 2012).
HT-29 subpopulations, some of which were mucus secreting These in vitro models enable the simple and reproducible
clones morphologically analogous to intestinal goblet cells study of cell mechanisms at molecular level that may be dif-
(Augeron and Laboisse 1984). Later, when the focus shifted ficult to perform in vivo. Nowadays, semipermeable plastic
to the ability of HT-29 cells to adapt to metabolic stress filter inserts used as culture systems facilitate the observation
conditions, various populations of mucus-secreting cells of molecules moving across the cell monolayer (compart-
were isolated including HT29-FU, HT29-18N2 and HT29- mentalized systems). Accordingly, in recent years, many
MTX (Huet et al. 1987; Lesuffleur et al. 1993). The stable studies have been performed to characterize the absorption,
clone HT29-MTX was isolated from the HT-29 cell resist- transport and bioavailability of food components or mole-
ance (irreversible) to high concentrations of Methatrexate cules of therapeutic interest. Even if caution should be exer-
(MTX), after a gradual adaptation to this anticancer drug. cised when comparing in vitro and in vivo results, these
This clone is capable of spontaneously differentiating into research works provide interesting information.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 5

Bioactive compounds: transport and demonstrate the antioxidant and anti-inflammatory potential
antioxidant/anti-inflammatory potential in of the polyphenols usually present in fruit and vegetables. The
intestinal cell culture models transport (Dhuique-Mayer et al. 2007; During et al. 2002) and
anti-inflammatory effect (Kaulmann et al. 2012) of carotenoids
Bioactive compounds like phytochemicals such as polyphe-
have also been studied using cell culture models.
nols and carotenoids, present interesting benefits for health
As already mentioned, cell monocultures have been
particularly due to their anti-inflammatory and anti-oxidant
extensively used to assess immune response modulation,
potential. Intestinal cell culture models have been extensively
antioxidant and anti-inflammatory effects of different com-
used to study these properties and their absorption and
ponents of fruits, vegetables and other natural products such
transport to assess their bioavailability and regulation of
as human breast milk lipids or phytic acid (Barrera and
intestinal permeability.
Sanchez 2015; Hollebeeck et al. 2012; Kang et al. 2009;
For example, a Cocoa flavan-3-ols absorption/secretion
Sergent et al. 2010; WeR glarz et al. 2007; Wojtal et al. 2013;
study was conducted by Kosi nska and Andlauer (2012)
Xu et al. 2012). Shimizu (2010) reviewed the interaction of
using Caco-2 cell monolayers. Their results suggest a para-
food components with the intestinal epithelium using the
cellular pathway of transport for the cathechin, epicatechin
Caco-2 line as cell model, particularly in studies of transport
and procyanidin B2. In addition, these authors concluded
pathways, modulation of transporter functions, TJ perme-
that compounds of a purified extract of cocoa powder might
ability and immune system regulation (Shimizu 2010).
be able to increase permeability with no harmful effects on
It should be also pointed out that food additives, adju-
cells, thereby increasing the bioavailability of flavan-3-ols
vants or food matrices may influence the intestinal transport
(Kosi nska and Andlauer 2012). Recently, Denis et al. (2015)
and bioavailability of these bioactive compounds (Nerurkar,
provided evidence for the capacity of cranberry phenolic
Burton, and Borchardt 1996) or have a direct influence on
fractions to reduce intestinal oxidative stress and inflamma-
intestinal function (Jiang et al. 2013). Recently, the inter-
tion while improving mitochondrial dysfunction using the
cell clone Caco-2/15 as a model (Denis et al. 2015). action of nanomaterials employed in the food industry with
Similarly, the results obtained by Bitzer et al. (2015) in a intestinal models, including cell cultures, was reviewed by
study of cocoa procyanidin fractions in Caco-2 and HT29 Lefebvre (Lefebvre et al. 2015). The migration of food con-
cell lines suggest that high-molecular-weight polymeric pro- tact materials like silver nanoparticles (AgNPs) from utensils
cyanidins are highly effective at preserving membrane integ- and storage containers was studied using the T84 cell line,
rity (in Caco-2 cells) and reducing inflammation markers as well as its impact on intestinal permeability and barrier
such as IL-8 (in HT29 cells) (Bitzer et al. 2015). In 2017, function. These authors concluded that small-size AgNPs
Martins et al. published a study on the antioxidant and anti- may increase intestinal permeability even in the absence of
inflammatory effects of grape pomace (GP), which is rich in any toxic effects on cells (Williams et al. 2016).
polymeric and glycosidic polyphenols. These authors showed Cell models have been useful not only for the study of food
a reduction in inflammatory markers (IL-8, PGE2) on IL-1b components but also for pharmaceutical research. Studies in
stimulated Caco-2 monolayers after the addition of tannase this field aim to evaluate potential toxic effects of drug candi-
(tannin acyl hydrolase) to the cultures, suggesting an dates and to improve the absorption and bioavailability of
improvement in the antioxidant and anti-inflammatory drugs, nanoparticles or other components used in drug deliv-
activities of GP polyphenol extracts (Martins et al. 2017). ery. For instance, in the case of the improvement of oral deliv-
The potential health benefits of polyphenol have been also ery of poorly permeable drugs, Maher et al. (2016) reviewed
studied by other researchers. The positive effect of kaempferol more than 200 intestinal permeation enhancers (PEs) that have
on the integrity of the TJ barrier was investigated using Caco- been tested in preclinical intestinal delivery models, including
2/HTB-37 cells and showed improvement in TJ protein assem- Caco-2 cells. These permeation enhancers alter intestinal per-
bly after flavonoid administration (Suzuki, Tanabe, and Hara meability in two different ways. First generation paracellular
2011). Likewise, Rodrıguez-Ramiro et al. (2013) studied the PEs target cell signaling pathways involved in disbandment of
molecular mechanism underlying the anti-inflammatory activ- TJs and the second generation can directly target the physical
ity of polyphenols and concluded that cocoa polyphenols disruption of TJs by interfering in intercellular hemophilic
effectively down-regulate the levels of inflammatory markers interactions. Transcellular PEs, act via modification of the
in vitro (Caco-2 cells) and in vivo (rat model) (Rodrıguez- integrity of the cell plasma membrane or via hydrophobisation
Ramiro et al. 2013). A flavonoid-rich concentrate from of the target therapeutic peptide. Maher et al. (2016) concluded
Opuntia ficus indica juice, a species of cactus, was evaluated by that models like epithelial monolayer cultures enable the detec-
Matias et al. (2014). Their results showed that co-incubation tion of PEs that modify barrier integrity and improve the bio-
of the concentrated with a stress-inductor attenuates the pro- availability of peptides even though such systems do not
duction of radicals significantly more than pre-incubation. consider drug formulation (Maher, Mrsny, and Brayden 2016).
This suggests that some of the main compounds present in the
concentrate, e.g. isorhamnetins derivatives that present low
Intestinal and immunological cells Mono- and
bioavailability, inhibit the formation of reactive oxygen species
co-cultures as gut inflammation models
(ROS) in the environment of epithelial intestinal cells. A simi-
lar positive response to inflammatory activity was observed To improve on simple cellular models, researchers developed
during co-incubation (Matias et al. 2014). All these studies 2D co-cultures as gut inflammation models (summarized in
6 
M. C. PONCE DE LEON-RODRIGUEZ ET AL.

Table 1). These models were established using the associ- microbial ligands involving Toll-like receptors (TLRs)
ation of intestinal and immune cells, the second most (Berghaus et al. 2010). However, these authors concluded
numerous type of cells present in intestinal epithelium, and that caution is required when interpreting and extrapolating
compartmentalized systems largely employed to grow intes- results due to possible changes in RAW cells in continuous
tinal cell lines, as mentioned above. Since the late 1990’s, culture (Berghaus et al. 2010). Moreover, possible inter-spe-
researchers have shown increasing interest in improving cies cross-reactions should also be taken into account in co-
intestinal mucosal barrier models by including immune cells culture models that combine murine (e.g. RAW cells) and
in the cultures (Kerneis et al. 2000). The main immune cells human cell lines as Caco-2 cells to study anti-inflamma-
used to establish these co-culture systems with intestinal cell tory effects.
lines include primary cells like human peripheral blood
mononuclear cells (PBMC) and macrophage cell lines of 2D co-culture models. Establishing co-cultures of different
either human (THP-1) or murine origin (RAW 264.7). cell types enables the study of cell-cell (intercellular) com-
munication and the investigation of the influence of these
interactions on growth, differentiation and immune
Pbmc
response. This also makes it possible to explore the effect of
PBMC are obtained from the buffy coats of healthy donors food components and the response of cells to exposure to
frequently through Ficoll gradient centrifugation and include the components in either healthy or pathological conditions.
lymphocytes, monocytes and dendritic cells. The presence of Even though in vitro models do not fully mimic the com-
cell markers for macrophages and dendritic cells has been plexity of in vivo physiology, these models facilitate the
described in co-cultures using PBMC (Leonard, Collnot, and study of cell contact mechanisms and responses to soluble
Lehr 2010). However, given the differences in their pheno- factors such as cytokines. Nowadays, the majority of the co-
type, cells isolated from blood might have different culture models involving intestinal and immune cells use
responses from those present in the intestinal lamina propria standard insert systems in which intestinal cells (upper com-
as they present (Kleiveland 2015). partment) are not in direct contact with the immunological
ones (lower compartment). However, there have been a few
THP1 cell line studies in which both intestinal and immune cells are seeded
The THP-1 cell line was isolated from the peripheral blood in the same compartment (Susewind et al. 2016) or using
of a 1-year old male patient suffering from acute monocytic inverted models (Ara ujo et al. 2016; des Rieux et al. 2007;
leukemia and characterized as a monocytic cell line Mori, Satsu, and Shimizu 2003; Tyrer et al. 2011). This
(Tsuchiya et al. 1980). These cells can be cultured in vitro allows direct contact between the two cell types and hence
for about 3 months (passage 25) with no modification in cell the study of migration of the immune cells into the intes-
sensitivity or activity. THP-1 cells can differentiate into mac- tinal monolayer (see particular features in Table-1).
rophages after phorbol-12-myristate-13-acetate (PMA) treat- However, 2D cell culture models have some limitations
ment or into dendritic cells by transferring them into including reduced cell-cell interactions, lack of cell-matrix
serum-free medium, and subsequently treating them with a interactions, and the tissue architecture is not completely
mixture of IL-4, GM-CSF, TNF-a and ionomycin (Chanput, recreated; Fitzgerald et al. (2015) reviewed these disadvan-
Peters, and Wichers 2015). This cell line has been character- tages in terms of evaluating new drug delivery strategies
ized as a model for immune modulation (Chanput et al. (Fitzgerald et al. 2015). Despite their limitations, 2D in vitro
2013; Chanput, Mes, and Wichers 2014) and has also been models are a useful tool to study the interactions between
widely used to study ROS production and inflammation. For food components and intestinal cells during intestinal
instance, researchers have employed THP-1 cell line as a inflammation. To date, only a few studies have linked the
model to analyze the anti-inflammatory effects of polyphe- effects of food components to gut inflammation using 2D
nols, (Chanput et al. 2010; Huang, Wu, and Yen 2006; Wu co-culture models. In this context, Caco-2 cells have been
et al. 2011) or fatty acids (Huang et al. 2012). co-cultivated with different immune cells in an attempt to
The use of THP-1 could have certain advantages over mimic the presence of the latter in the lamina propria
PBMC for example, due to its homogeneous genetic back- beneath the intestinal epithelium. For example, the Caco-2
ground that reduces the variability of the cell phenotype, cell line has been associated with THP-1 cells to study
thereby improving the reproducibility of the experiments. effects of lycopene, a carotenoid present in red fruits and
vegetables, on pro-inflammatory targets. The authors of that
study found that a low concentration (2 mM) of lycopene
RAW 264.7 cell line reinforced pro-inflammatory gene expression in THP-1 cells,
Finally, a monocytic murine cell line (RAW 264.7) has also whereas higher concentrations (5–20 mM) had an anti-
been frequently used in co-cultures with Caco-2 cells. The inflammatory effect with a decrease in ROS (Makon-
RAW 264.7 cell line was established from a murine tumor Sebastien et al. 2014). Other authors associated Caco-2 cells
induced by the Abelson leukemia virus and presents macro- with the RAW 264.7 cell line to study the anti-inflammatory
phage properties (Raschke et al. 1978). It has been shown effects of natural extracts. In one study, researchers observed
that these cells mimic murine bone marrow-derived macro- anti-inflammatory properties of a fruit juice enriched with
phages in terms of cell surface receptors and response to pine bark extract which decreased after digestion (Frontela-
Table 1. Gut inflammation 2D co-culture models.
Model Cell type(s) & source Objective(s) Particular features Reference
Caco-2/THP1 Human colon adenocarcinoma / Effect of lycopene on pro-inflammatory targets of intes- / (Makon-Sebastien et al. 2014)
Human peripheral blood (acute tinal and immune cell lines
monocytic leukemia) cell lines Microparticle uptake / (Moyes, Morris, and Carr 2010)
Effect of macrophages on intestinal epithelial cells / (Satsu et al. 2006)
Evaluation of the cross talk between intestinal epithe- / (Kanzato, Manabe, and
lium and macrophages Shimizu 2001)
Migration of immune cells into intestinal epithelial Caco-2 cells were cultured on inverted (Mori, Satsu, and Shimizu 2003)
cell monolayers Millicell inserts (12 mm pore size) and
then re-orientated before adding THP-1
to allow contact with the porous mem-
brane at the basolateral side of the cells.
Effect of chickpea protein hydrolysates on cell / on-Calle, Alaiz, and
(Gir
proliferation Vioque 2010)
Bioaccessibility, bioavailability and anti-inflammatory / (Zhang et al. 2017)
effects of polyphenols
Caco-2/Raw 264.7 Human colon adenocarcinoma / Anti-inflammatory effects of hit or lead drugs from the Compounds were chosen according to a (Galvez-Llompart et al. 2013)
Murine macrophage cell lines MicroSource Pure Natural Products Collection and quantitative structure-activity relationship
Sigma-Aldrich database model based on molecular topology to
predict the IL-6 mediated anti-UC activity.
Effect of fucoidan (from brown algae) on gut / (Tanoue et al. 2008)
inflammation
Anti-inflammatory effects of Smabucus nigra fruit extract / (Olejnik et al. 2015)
on LPS-stimulated macrophage-type cells
Anti-inflammatory effects of Sasa quelpaertensis leaf / (Kim et al. 2015)
extract (SQE) on LPS-stimulated co-culture
Anti-inflammatory capacity of fruit juices enriched with / (Frontela-Saseta et al. 2013)
pine bark extract (PBE)
Caco-2/ PBMC Human colon adenocarcinoma Effect of arsenical species on the release of cytokines / (Calatayud et al. 2015)
cell line / Human peripheral Lactobacillus casei subsp. rhamnosus (Lcr35) anti-inflam- / (Fang et al. 2010)
blood mononuclear cells matory effect on Caco-2 cells.
Effect of bacterial products (LPS, MDP) on enterocyte- Caco-2 cells were cultured on inverted (Tyrer et al. 2011)
macrophage interaction Transwells and then re-orientated before
adding PBMC to allow cell-cell contact.
Interaction between PBMC and enterocytes during chal- / (Parlesak et al. 2004)
lenge with bacteria (Nonpathogenic E. coli)
Human colon adenocarcinoma Validation of an in vitro M-cell model Caco-2 cells were cultured on inverted (Tyrer et al. 2002)
cell line / Murine peripheral Transwells (0.3 mm pore size) and then
blood mononuclear cells re-orientated before adding isolated
Peyer’s patch lymphocytes to allow cell-
cell contact.
Caco-2/HT29-MTX/PBMC Human colon adenocarcinoma Salmonella enterica and intestinal cells interactions Intestinal cell lines were seeded on the (Dostal et al. 2014)
cell lines / Human peripheral under different Fe concentrations using an in vitro apical side of the inserts to which S.
blood mononuclear cells colonic fermentation system inoculated with immobi- enterica N-15 in DMEM or fermentation
lized child microbiota effluents, both with different Fe concen-
trations, was added. Freshly collected
PBMC were added to the basolateral com-
partment with different concentrations
of Fe.
Caco-2/HT29-MTX/THP-1 Human colon adenocarcinoma / Anti-inflammatory and antioxidant capability of carote- Intestinal cell lines were seeded on inserts (Kaulmann, Legay, et al. 2016)
Human peripheral blood noids/polyphenols from plum and cabbage digesta apical side and immune cells on the baso-
(acute monocytic leukemia) on a stimulated model (LPS, TNF-a & IL-1b) lateral compartment.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION

cell lines
(Continued)
7
Table 1. Continued. 8
Model Cell type(s) & source Objective(s) Particular features Reference
Caco-2/Raji B Human colon adenocarcinoma / Improvement of an in vitro model of human follicle- Two models were set up, one with Raji B (des Rieux et al. 2007)
Human Burkitt’s lymphoma associated epithelium (FAE) to study nanoparticle cells in the basolateral compartment of a
cell lines transport mechanisms by M cells standard compartmentalized model and
the other where inserts containing Caco-2
cells were inverted in order to seed Raji B
cells in contact with the porous mem-
brane at the basolateral side of the intes-


tinal cells.
Caco-2/HT29-MTX/Raji B Human colon adenocarcinoma / Establishment of a tissue-engineered model for oral Two models were set up: one in which (Antunes et al. 2013; Araujo
Human Burkitt’s lymphoma drug delivery studies Intestinal cell lines were seeded on et al. 2016; Ara
ujo and
M. C. PONCE DE LEON-RODR

cell lines Transwell inserts and Raji-B cells were Sarmento 2013)
added to the basolateral compartment
and an inverted model in which intestinal
cells were seeded on the basal side of
the membrane and then Raji B cells were
IGUEZ ET AL.

seeded on the apical side of the insert.


Development of a triple culture intestinal model to Intestinal cell lines were seeded on the (Schimpel et al. 2014)
study nanoparticle mobility apical side of Transwell inserts and Raji-B
cells were seeded on the basolateral
compartment.
Caco-2/ISO-HAS-1 Human colon adenocarcinoma/ Evaluation of co-culture interactions with nanosized Transwell inserts were coated with collagen I (Kasper et al. 2016)
Human microvascular endo- drug vehicles or contrast agents during an inflamma- and inverted, ISO-HAS-1 cells were seeded
thelial cell lines tory induced state on the lower side and after 2 h of adhe-
sion, the inserts were re-inverted and
Caco-2 cells seeded in the upper
compartment.
SW480/THP-1 Human colorectal adenocarcin- Investigation of the effects of HMGB1 inhibitors in a SW480 cells were prepared in the lower (Wang 2015)
oma / Human peripheral LPS stimulated model chamber of Transwell inserts and THP-1
blood (acute monocytic leu- cells were loaded into the
kemia) cell lines upper chamber.
m-ICd2 / DCs Murine intestinal epithelial cells/ Probiotic bacteria strain-specific immune-modulatory / (Zoumpopoulou et al. 2009)
dendritic cells / murine bone capacities: impact on dendritic cells maturation
marrow-DCs cells
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 9

Saseta et al. 2013). Furthermore, it has been shown in other PBMC as immune cells embedded in a collagen layer. The
studies that fruit (e.g. eldelberry) and leaf (e.g. bamboo) model was used to evaluate the efficacy of different formula-
extracts down-regulated the expression of pro-inflammatory tions of an anti-inflammatory drug (budesonide) used in the
cytokine (IL-1b, IL-6, TNF-a) and antioxidant enzymes treatment of IBD. Their results show the ability of the model
(iNOS, COX) genes and/or suppressed their secretion (Kim to differentiate therapeutic efficacy among different formula-
et al. 2015; Olejnik et al. 2015). Recently, Kaulmann et al. tions while adequately representing the complex patho-
(2016) associated two intestinal cell lines (Caco-2/HT29 physiological changes observed in vivo (Leonard et al. 2012;
MTX) with THP-1 cells to study the anti-inflammatory and Leonard, Collnot, and Lehr 2010). Li et al. (2013) also devel-
antioxidant capability of plum and cabbage digesta. Their oped a 3D intestinal model involving intestinal cells (Caco-2,
results revealed a reduction in certain markers of inflamma- HT29MTX), fibroblast-type cells and macrophage-type cells
tion (IL-6, IL-8 and NF-jB) and oxidative stress (Nrf2), to study drug absorption. The authors concluded that their
although the effects were moderate and could not be aligned model mimics the native intestinal epithelial layer more
to either cabbages or plums, nor to carotenoids or polyphe- closely. They also demonstrated an improved correlation
nols (Kaulmann, Legay, et al. 2016). between the absorptive permeability across the 3D co-culture
model and the human absorption fraction in the case of
3D (co)-culture models. In vitro models are constantly moderate and highly permeable drugs (Li et al. 2013).
evolving from simple monocultures, through 2D (co-) cul- More recently, Wu et al. (2017) assessed the toxicity and
tures using plastic supports, to 3D cultures in which cells inflammatory effects of ZnO nanoparticles (ZnO NPs) using
are grown within an extracellular matrix (ECM). Kim et al. 2D and 3D Caco-2 cell culture models. ZnO NPs have been
(2014) observed differences in barrier function (permeabil- used in dietary supplements and in food additives and food
ity) between a 2D and 3D villi model using Caco-2 cells (S. packaging materials because of their antimicrobial activity.
H. Kim et al. 2014). Similarly, Yu et al. (2012) reported var- Wu et al. results showed that ZnO NPs induced the toxicity
iations during cell differentiation; according to their experi- of 2D and 3D cultures in different size-dependent manners.
ments, 3D models could be more time efficient with a more Their results also indicated that ZnO NPs induced a chronic
and realistic cell differentiation (Yu et al. 2012). Another of inflammatory response in 2D cell culture and an acute
the limitations of 2D models, particularly in tumor research, inflammatory response in 3D cell culture, indicating that
is the absence of a true ECM with changes in structure, cell space organization have an important impact on cell
adhesive potential, mechanotransduction, and results of responses (Wu et al. 2017). Even if today, most research
exposure to soluble factors (Ryan et al. 2016). In fact, the using 3D models has pharmaceutical goals, in the food field,
ECM gives tissues mechanical properties and favors cell such models could be used to study absorption and the anti-
communication. The anchorage of membrane receptors to oxidant and/or anti-inflammatory properties of bioactive
the ECM regulates the cells’ interpretation of signals from food compounds during intestinal inflammation.
their environment (Abbott 2003). Several biochips have been established that are thought to
Thanks to advances bioengineering, 3D co-culture sys- be more representative of the in vivo microenvironment try-
tems have been designed using different platforms like scaf- ing to mimic intestinal functions. The development of these
fold constructions, hydrogels (e.g. collagens, laminins), complex devices, passing from static to fluidic cultures,
spheroids and microfluidic devices with channels that enable opens new opportunities to understand organ-level interac-
controlled diffusion of nutrients (Fitzgerald et al. 2015). The tions. In the food field, they could be employed to study
aim of 3D models and the frequently termed organs- nutrient absorption and the impact of food components on
on-chips or biochips (microfluidic devices), is to provide a immune modulation. For instance, a new integrated micro-
cellular microenvironment that brings in vitro conditions fluidic platform, called Nutrichip, has been developed to
closer to the physiological reality of the organization and investigate the potential of the immune modulatory function
dynamic properties of tissues (summarized in Table 2). For of dairy foods (Ramadan et al. 2013; Vergeres et al. 2012).
example, Juuti-Uusitalo et al. (2011) employed 3D models This system consists of a monolayer of epithelial cells inter-
(T84 and Caco-2 cells) to study the effect of TNF (TNFSF2) acting with immune cells mounted on a chip holder device.
and INFc on intestinal epithelial morphogenesis and perme- Different perfusion microchannels allow a uniform radial
ability. Their work revealed differences in the response to fluid flow of the culture medium and an inflammatory
stimuli using a 3D model, and showed that apoptosis is an stimulus or a digesta; cytokine production is quantified
important mechanism in increased permeability after TNF using an on-chip immunomagnetic assay. More recently, a
stimulation. They also demonstrated that this pro-inflamma- similar miniaturized Caco-2/U937 co-culture model was
tory cytokine inhibits normal intestinal epithelial morpho- used to characterize TJ disruption and immune response
genesis resulting in the formation of ‘leaky’ monolayers in modulation at intestinal level (Ramadan and Jing 2016).
the case of T84 3D model (Juuti-Uusitalo et al. 2011). More research is still needed to completely control all the
To our knowledge, most of these 3D and microfluidic model features (Pampaloni, Reynaud, and Stelzer 2007; Ravi
models have been applied in the pharmaceutical field with a et al. 2015). For example, ECM composition and stiffness
focus on drug toxicology and screening. For instance, (Langhans 2018; Wallace and Rosenblatt 2003), gas
Leonard et al. (2010, 2012), established an inflamed 3D exchange, chemical and mechanical signal control and inter-
model using the intestinal cell line Caco-2 (C2Bbe1) and stitial flows have a crucial impact on cell behavior and
Table 2. Gut 3D culture models. 10

Model Cell type(s) & source Objective(s) Particular features Reference


HEALTHY GUT D human small intes- Caco-2 (human colon Development of a 3D hydrogel scaffold Cells cultured on a soft-hydrogel structure. (Yu et al. 2012)
tinal villous adenocarcinoma) model to accurately replicate the shape
and size of human small intestinal villi to
test drug permeability
Multicellular organotypic HCT-8 (human ilieocecal adenocarcin- Development of an organotypic model with Model includes fibroblasts, lymphocytes, epi- (Salerno-Goncalves,
model of human intes- oma), CCD-18Co (human colon close structural and functional resem- thelial and endothelial cells embedded in Fasano, and
tinal mucosa fibroblast), HUVEC (human umbil- blance to the human intestinal mucosa a protein enriched collagen I matrix. Cells Sztein 2011)
ical vein endothelial cells) can differentiate into several lineages


(goblet cells, M cells and differentiated


enterocytes).
3D setup of intes- Caco-2 (human colon adenocarcin- Development of a 3D intestinal model for Immune cells were embedded in a collagen (Collnot, Susewind, and
M. C. PONCE DE LEON-RODR

tinal mucosa oma), THP-1 (human macro- drug delivery studies and to assess nano- scaffold, seeded on the apical side of Lehr 2013; Susewind
phages), MUTZ-3 (human material toxicity in healthy or dis- inserts and Caco-2 cells, on top of et al. 2015)
dendritic cells) ease conditions this layer.
3D co-culture (intestinal Caco-2 (human colon adenocarcin- To study TJ expression and cell differenti- Co-cultures of intestinal and endothelial (Matsusaki et al. 2015)
IGUEZ ET AL.

epithelial cells-fibro- oma) / NHDF (normal human der- ation in a 3D model cells; 1L-Caco-2/1L-NHDF and 1L-Caco-2/
blast model) mal fibroblasts) 8L-NHDF were constructed using a cell-
coat (nano-ECM of fibronectin and gel-
atin) technology.
3D intestinal culture T84 (human colon carcinoma, lung Effect of TNFa and IFNc on paracellular per- Cells were seeded in MatrigelTM in order to (Juuti-Uusitalo et al. 2011)
metastasis), Caco-2 (human colon meability and morphogenesis in 3D T84 form spheres.
adenocarcinoma) and Caco-2 luminal spheres
Caco-2 (human colon Development of microporous, polymeric Membranes can be integrated with micro- (Esch et al. 2012)
adenocarcinoma) membranes that are either flat or contain fluidic, multi-organ cell culture systems,
controllable 3-dimensional shapes that providing access to both apical and baso-
can be integrated in microfluidic, multi- lateral sides.
organ cell culture systems
Caco-2 (human colon Study of epithelial morphogenesis To produce cysts, Caco-2 cells were plated (Jaffe et al. 2008)
adenocarcinoma) either on top of the ECM (MatrigelTM), for
time lapse or embedded in the ECM (col-
lagen I and MatrigelTM), for
immunofluorescence.
Caco-2 (human colon Toxicity and inflammatory effects of differ- The cells were seeded and immobilized in (Wu et al. 2017)
adenocarcinoma) ent-sized ZnO nanoparticles (NPs) at vari- agarose gel.
ous concentrations seeded in 3D cultures
Human gut-on-a-chip Caco-2 (human colon Development of a biomimetic ‘human gut- Micro-device composed of two microfluidic (Kim et al. 2012; Kim and
adenocarcinoma) on-a-chip’ channels separated by a porous flexible Ingber 2013)
membrane coated with ECM (collagen
and MatrigelTM) and lined with human
intestinal epithelial (Caco-2) cells.
Intestinal microflui- Caco-2 (human colon Development of a microchip-based system Microchip composed of a glass slide, a per- (Imura et al. 2009)
dic system adenocarcinoma) to mimic the intestine meable membrane, and polydimethylsi-
loxane sheets, containing microchannels
and Caco-2 cells seeded on
the membrane.
Intestinal microscale cell Caco-2 and HT29-MTX (human Development of an in vitro microscale cell Intestinal cells seeded on polycarbonate (0.4 (Mahler, Shuler, and
culture analog (mCCA) adenocarcinoma), HepG2/C3A culture analog (mCCA) to the gastrointes- mm) membranes, mCCA coated with poly- Glahn 2009)
(human hepatocellular carcinoma) tinal tract D-lysine and plasma fibronectin and
HepG2/C3A cells seeded into the liver
chamber. A peristaltic pump was used to
simulate medium re-circulation.
(Continued)
Table 2. Continued.
Model Cell type(s) & source Objective(s) Particular features Reference
Intestinal Caco-2 (human colon Development of an integrated micro- The microfluidic structure is divided into two (Kimura et al. 2008)
microfluidic adenocarcinoma) fluidic system for long-term perfu- independent channels separated by a
device sion culture and on-line semipermeable membrane enabling the
monitoring of intestinal tis- detection of polarized Caco-2 trans-
sue models port activity.
Microfluidic Caco-2 (human colon Reproduce the 3D villi structure and Cells were seeded on a collagen scaffold (Shim et al. 2017)
gut-on- adenocarcinoma) the fluidic shear in a microflui- (villi) incorporated into a microfluidic
a-chip dic chip device consisting of three layers of polydi-
methyl-siloxane (PDMS), a slide glass and
a polyester (PET) membrane
Intestinal Caco-2 (human colon Evaluation of Ca2þ transport The chip was made of two PDMS layers, two (Huang et al. 2014)
microfluidic adenocarcinoma) polymethylmethacrylate (PMMA) layers, a
chip PET membrane (0.4mm) and a glass slide.
Ag/AgCl electrodes were included in
the device.
Intestinal Caco-2 (human colon Development of a microfluidic device Caco-2 cells were seeded on a porous mem- (Chi et al. 2015)
microfluidic adenocarcinoma that mimics human intes- brane coated with fibronectin between
device tinal properties two layers of polydimethylsilox-
ane (PDMS).
INFLAMED GUT 3D in vitro intestinal Caco-2 and HT29-MTX (human Development of an improved 3D in vitro MEFs were dispersed in a type rat tail colla- (Li et al. 2013)
mucosa model adenocarcinoma), THP-1 (human intestinal mucosa model to evaluate gen solution. Intestinal cells were seeded
peripheral blood -acute monocytic drug absorption. on the top of the formed collagen gel.
leukemia), MEFs, P0 (primary The co-culture was then transferred to a
mouse embryonic fibroblast) receiver plate pre-seeded with THP-1
derived macrophages.
Intestinal inflamed 3D Caco-2, HT29, T84 (human colon Development of a 3D co-culture of human Macrophages and dendritic cells derived (Leonard et al. 2012;
cell-culture model adenocarcinoma) / PBMC (human intestinal and immune cells stimulated from PBMC were embedded in a collagen Leonard, Collnot, and
peripheral blood mono- model for anti-inflammatory drug screen- layer on a Transwell insert and Caco-2 Lehr 2010)
nuclear cells) ing and their formulations cells were seeded on top.
Caco-2 (human adenocarcinoma), Evaluation of the delivery efficacy of THP-1 and Caco-2 cells were embedded in a (Huang et al. 2014)
THP-1 (human peripheral blood nanoparticles Matrigel TM solution.
-acute monocytic leukemia)
3D co-culture (intestinal HT29 (human colon adenocarcin- Study of Salmonella enterica coloniza- U937 cells were activated upon collagen- (Barrila et al. 2017)
epithelial cell-macro- oma), U937 (human monocyte- tion patterns coated scaffolds. HT-29 epithelial cells
phage) model histiocytic lymphoma) were then added and the 3D model was
cultured in the NASA Rotating Wall Vessel
bioreactor until optimal differentiation
was reached.
NutriChip Caco-2 (human colon adenocarcin- Development of an integrated microfluidic The core component of the NutriChip is a (Ramadan et al. 2013;
oma), U937 (human monocyte- platform to investigate the potential miniaturized artificial human gastrointes- Vergeres et al. 2012)
histiocytic lymphoma) immuno-modulatory function of tinal tract (GIT), which consists of a con-
dairy food fluent layer of epithelial cells separated
from a co-culture of immune cells by a
permeable membrane.
Microfluidic chip Caco-2 (human adenocarcinoma), Development of a microfluidic-based Continuous perfusion of culture media from (Ramadan and Jing 2016)
U937 (human monocyte- histio- dynamic in vitro model of human intes- the apical and basolateral side of the por-
cytic lymphoma) tinal barrier ous membrane mimics physiological flow.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION
11
12 
M. C. PONCE DE LEON-RODRIGUEZ ET AL.

responses (Griffith and Swartz 2006). Other challenges asso- human epithelial cells may be in the way to be overcome
ciated with the use of these kinds of models concern the even if further improvements are required to optimize
impact of seeding density on cell responses and develop- these models.
ment, as it has been shown for other type of tissues (Dvir-
Ginzberg et al. 2003; Holy, Shoichet, and Davies 2000;
Conclusion and perspectives
Ohnuma, Arkin, and Holland 1986; Watt 1988).
Optimization of these models is thus required to be able to Functional in vitro models of the gut have an essential role
correlate 3D model and in vivo results. in the assessment of the risks or benefits of food as an alter-
Finally, it should be pointed out that recent advances in native to animal studies. Despite the advances in cell culture
human intestinal epithelial stem cell culture methods, such models, much remains to be achieved in the development of
as the discovery of the growth factors required for stem cells complex gut inflammation models. From simple intestinal
homeostasis, have allowed the generation of long-term cul- monocultures, through co-cultures to 3D cultures within an
ture systems: human intestinal organoids (HIOs) and enter- ECM, in vitro models of the intestine have gain considerable
oids/colonoids (HIEs). These ex vivo and in vitro models importance as a tool to create more physiologically appro-
reproduce physiological characteristics of the human intes- priate conditions. Most inflamed gut models are currently
tine in vivo (Zachos et al. 2015). HIOs are established in run in 2D conditions to study interactions between the gut
vitro using induced pluripotent stem cells (iPSCs) or embry- and bioactive food compounds or drugs. In addition, 3D
onic stem cells (ESCs) (Spence et al. 2011; McCracken et al. systems including microfluidic devices, HIOs and HIEs are a
2011). HIEs are developed from dissociated crypts/stem cells promising way to understand complex whole-organ human
from intestinal tissue (Sato et al. 2011; Saxena et al. 2015). physiology, and are a good alternative to in vivo studies.
In contrast to other culture techniques that contain cells of Major challenges such as the characterization and validation
mesenchymal origin, HIEs allow the study of isolated epithe- of such models as gut inflammation models will concern
lial cells (VanDussen et al. 2014). HIEs models are cultivated how to integrate these technologies in the drug and food
in monolayers or in 3D forms (spheroids) using Matrigel as fields. Although caution should be exercised when compar-
ECM (VanDussen et al. 2014; Costantini et al. 2018). These ing in vitro and in vivo results, such research should provide
models include several differentiated cell types (enterocytes, interesting information.
goblet, enteroendocrine and Paneth cells) and could be
established from different segments of the intestine which
References
could help to better understand the specific-region functions
(Middendorp et al. 2014). HIEs models are derived of non- Abbott, A. 2003. Biology’s new dimension. Nature 424(6951):870.
transformed small intestine and colonic tissues established doi:10.1038/424870a.
Abraham, C., and J. H. Cho. 2009. Inflammatory bowel disease. New
from individuals biopsies (VanDussen et al. 2014) or from England Journal of Medicine 361(21):2066–78. doi:10.1056/
surgically resected intestinal tissues (Sato et al. 2011). This NEJMra0804647.
could be considered as an inconvenient due to ethical issues, Al-Sadi, R., D. Ye, H. M. Said, and T. Y. Ma. 2011. Cellular and molecu-
however it can also be seen as an advantage because it could lar mechanism of interleukin-1b modulation of caco-2 intestinal epi-
simplify the study of individual variations in human epithe- thelial tight junction barrier. Journal of Cellular and Molecular
Medicine 15(4):970–82. doi:10.1111/j.1582-4934.2010.01065.x.
lial function. Especially host-pathogen interactions with Alzamora, R., F. O’Mahony, W.-H. Ko, T. W.-N. Yip, D. Carter, M.
potential use in patient-specific assays with repeatability Irnaten, and B. J. Harvey. 2011. Berberine reduces cAMP-Induced
already observed by VanDussen et al. (2014). These models Chloride secretion in T84 human colonic carcinoma cells through
also bring new opportunities, for instance, for the develop- inhibition of basolateral KCNQ1 channels. Frontiers in Physiology 2:
33. doi:10.3389/fphys.2011.00033.
ment of personalized treatments for chronic diseases.
Andoh, A.,. K. Kinoshita, I. Rosenberg, and D. K. Podolsky. 2001.
However, disadvantages of this type of cell culture systems Intestinal trefoil factor induces decay-accelerating factor expression
include the high cost (growth factors) and the fact that they and enhances the protective activities against complement activation
are labor intensive (Costantini et al. 2018). in intestinal epithelial cells. Journal of Immunology 167(7):3887–93.
Recently, researchers used HIEs models to study norovi- doi:10.4049/jimmunol.167.7.3887.
Antunes, F., F. Andrade, F. Ara ujo, D. Ferreira, and B. Sarmento. 2013.
rus and rotavirus (food/water spread). The authors con-
Establishment of a triple co-culture in vitro cell models to study
cluded that this type of models are suitable to study human intestinal absorption of peptide drugs. European Journal of
enteric virus pathogenesis. Previous attempts to study virus Pharmaceutics and Biopharmaceutics 83(3):427–35. doi:10.1016/
replication using other in vitro models have been unsuccess- j.ejpb.2012.10.003.
ful after many years because of the lack of a robust and Araujo, F., C. Pereira, J. Costa, C. Barrias, P. L. Granja, and B.
Sarmento. 2016. In vitro M-like cells genesis through a tissue-engi-
reproducible intestinal adapted cell culture system (Ettayebi neered triple-culture intestinal model. Journal of Biomedical
et al. 2016; Saxena et al. 2015, 2017; Costantini et al. 2018). Materials Research Part B: Applied Biomaterials 104(4):782–8.
Thus, organoids and enteroids/colonoids may be helpful in doi:10.1002/jbm.b.33508.
other areas of food research and beyond. For instance, the Araujo, F., and B. Sarmento. 2013. Towards the characterization of an
possibility to co-cultivate them with immune cells or micro- in vitro triple co-culture intestine cell model for permeability stud-
ies. International Journal of Pharmaceutics 458(1):128–34.
organisms would allow the generation of a more complex doi:10.1016/j.ijpharm.2013.10.003.
intestinal model in order to study intestinal inflammation. Arpaia, N., C. Campbell, X. Fan, S. Dikiy, J. van der Veeken, P.
Technical difficulties to extract and maintain viable primary deRoos, H. Liu, J. R. Cross, K. Pfeffer, P. J. Coffer, and A. Y.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 13

Rudensky. 2013. Metabolites produced by commensal bacteria pro- Calatayud, M., J. V. Gimeno-Alca~ niz, V. Devesa, and D. Velez. 2015.
mote peripheral regulatory T-cell generation. Nature Proinflammatory effect of trivalent arsenical species in a co-culture
504(7480):451–5. doi:10.1038/nature12726. of caco-2 cells and peripheral blood mononuclear cells. Archives of
Artursson, P., K. Palm, and K. Luthman. 2001. Caco-2 monolayers in Toxicology 89(4):555–64. doi:10.1007/s00204-014-1271-1.
experimental and theoretical predictions of drug transport1. Cammarota, G., G. Ianiro, R. Cianci, S. Bibb o, A. Gasbarrini, and D.
Advanced Drug Delivery Reviews 46(1-3):27–43. doi:10.1016/S0169- Curr o. 2015. The involvement of gut microbiota in inflammatory
409X(00)00128-9. bowel disease pathogenesis: Potential for therapy. Pharmacology &
Augeron, C., and C. L. Laboisse. 1984. Emergence of permanently dif- Therapeutics 149 :191–212. doi:10.1016/j.pharmthera.2014.12.006.
ferentiated cell clones in a human colonic cancer cell line in culture Cario, E., I. M. Rosenberg, S. L. Brandwein, P. L. Beck, H.-C.
after treatment with sodium butyrate. Cancer Research 44(9):3961–9. Reinecker, and D. K. Podolsky. 2000. Lipopolysaccharide activates
Barnett, A. M., N. C. Roy, W. C. McNabb, and A. L. Cookson. 2016. distinct signaling pathways in intestinal epithelial cell lines express-
Effect of a semi-purified oligosaccharide-enriched fraction from cap- ing toll-like receptors. Journal of Immunology 164(2):966–72.
rine milk on barrier integrity and mucin production of co-culture doi:10.4049/jimmunol.164.2.966.
models of the small and large intestinal epithelium. Nutrients Caro, I., X. Boulenc, M. Rousset, V. Meunier, M. Bourrie, B. Julian, H.
8(5):267. doi:10.3390/nu8050267. Joyeux, C. Roques, Y. Berger, A. Zweibaum, and G. Fabre. 1995.
Barrera, G. J., and G. Sanchez. 2015. Cytokine modulation (IL-6, IL-8, Characterisation of a newly isolated caco-2 clone (TC-7), as a model
IL-10) by human breast milk lipids on intestinal epithelial cells of transport processes and biotransformation of drugs. International
(caco-2). Journal of Maternal-Fetal and Neonatal Medicine 0(0):1–8. Journal of Pharmaceutics 116(2):147–58. doi:10.1016/0378-
doi:10.3109/14767058.2015.1091879. 5173(94)00280-I.
Barrila, J., Yang, J. Crabbe, A. Sarker, S. F. Liu, Y. Ott, C. M. Nelman- Chanput, W., J. Mes, R. A. M. Vreeburg, H. F. J. Savelkoul, and H. J.
Gonzalez, M. A. Clemett, S. J. Nydam, S. D. Forsyth. R. J. et al. Wichers. 2010. Transcription profiles of LPS-stimulated THP-1
2017. Three-dimensional organotypic co-culture model of intestinal monocytes and macrophages: A tool to study inflammation modu-
epithelial cells and macrophages to study Salmonella enterica colon- lating effects of food-derived compounds. Food & Function
ization patterns. Npj Microgravity 3(1):10. doi:10.1038/s41526-017- 1(3):254–61. doi:10.1039/C0FO00113A.
0011-2. Chanput, W., J. J. Mes, H. F. J. Savelkoul, and H. J. Wichers. 2013.
Beduneau, A., C. Tempesta, S. Fimbel, Y. Pellequer, V. Jannin, F. Characterization of polarized THP-1 macrophages and polarizing
Demarne, and A. Lamprecht. 2014. A tunable caco-2/HT29-MTX ability of LPS and food compounds. Food & Function 4(2):266–76.
co-culture model mimicking variable permeabilities of the human
doi:10.1039/C2FO30156C.
intestine obtained by an original seeding procedure. European Chanput, W., J. J. Mes, and H. J. Wichers. 2014. THP-1 cell line: An in
Journal of Pharmaceutics and Biopharmaceutics 87(2):290–8.
vitro cell model for immune modulation approach. International
doi:10.1016/j.ejpb.2014.03.017.
Immunopharmacology 23(1):37–45. doi:10.1016/j.intimp.2014.08.002.
Behrens, I., P. Stenberg, P. Artursson, and T. Kissel. 2001. Transport of
Chanput, W., V. Peters, and H. Wichers. 2015. THP-1 and U937 Cells.
lipophilic drug molecules in a new mucus-secreting cell culture
In The impact of food bioactives on health, eds. K. Verhoeckx, P.
model based on HT29-MTX cells. Pharmaceutical Research
Cotter, I. L opez-Exp osito, C. Kleiveland, T. Lea, A. Mackie, T.
18(8):1138–45. doi:10.1023/A:1010974909998.
Requena, D. Swiatecka, and H. Wichers, 147–159. New York, NY:
Beltran, A. R., Carraro-Lacroix, L. R. Bezerra, C. N. A. Cornejo, M.
Springer International Publishing.
Norambuena, K. Toledo, F. Araos, J. Pardo, F. Leiva, A. Sanhueza.
Chantret, I., A. Barbat, E. Dussaulx, M. G. Brattain, and A. Zweibaum.
C. et al. 2015. Escherichia coli heat-stable enterotoxin mediates naþ/
1988. Epithelial polarity, villin expression, and enterocytic differenti-
H þ exchanger 4 inhibition involving cAMP in T84 human intestinal
ation of cultured human colon carcinoma cells: A survey of twenty
epithelial cells. PLoS ONE 10(12):e0146042. doi:10.1371/
journal.pone.0146042. cell lines. Cancer Res 48(7):1936–42.
Benjamin, J., G. K. Makharia, M. Kalaivani, and Y. K. Joshi. 2008. Chantret, I., A. Rodolosse, A. Barbat, E. Dussaulx, E. Brot-Laroche, A.
Nutritional status of patients with Crohn’s disease. Indian Journal of Zweibaum, and M. Rousset. 1994. Differential expression of sucrase-
Gastroenterology: Official Journal of the Indian Society of isomaltase in clones isolated from early and late passages of the cell
Gastroenterology 27(5):195–200. line caco-2: Evidence for glucose-dependent negative regulation.
Berghaus, L. J., J. N. Moore, D. J. Hurley, M. L. Vandenplas, B. P. Journal of Cell Science 107(1):213–25.
Fortes, M. A. Wolfert, and G.-J. Boons. 2010. Innate immune Cheng, K.-C., C. Li, and A. S. Uss. 2008. Prediction of oral drug
responses of primary murine macrophage-lineage cells and RAW absorption in humans – from cultured cell lines and experimental
264.7 cells to ligands of toll-like receptors 2, 3, and 4. Comparative animals. Expert Opinion on Drug Metabolism & Toxicology
Immunology, Microbiology & Infectious Diseases 33(5):443–54. 4(5):581–90. doi:10.1517/17425255.4.5.581.
doi:10.1016/j.cimid.2009.07.001. Chi, M., B. Yi, S. Oh, D.-J. Park, J. H. Sung, and S. Park. 2015. A
Bitzer, Z. T., S. L. Glisan, M. R. Dorenkott, K. M. Goodrich, L. Ye, microfluidic cell culture device (lFCCD) to culture epithelial cells
S. F. O’Keefe, J. D. Lambert, and A. P. Neilson. 2015. Cocoa procya- with physiological and morphological properties that mimic those of
nidins with different degrees of polymerization possess distinct the human intestine. Biomedical Microdevices 17(3):58. doi:10.1007/
activities in models of colonic inflammation. The Journal of s10544-015-9966-5.
Nutritional Biochemistry 26(8):827–31. doi:10.1016/ Cho, J. A., and E. Park. 2015. Curcumin utilizes the anti-inflammatory
j.jnutbio.2015.02.007. response pathway to protect the intestine against bacterial invasion.
Bourgine, J., I. Billaut-Laden, M. Happillon, J.-M. Lo-Guidice, V. Nutrition Research and Practice 9(2):117–22. doi:10.4162/
Maunoury, M. Imbenotte, and F. Broly. 2012. Gene expression nrp.2015.9.2.117.
profiling of systems involved in the metabolism and the disposition Cho, J. H. 2008. The genetics and immunopathogenesis of inflamma-
of xenobiotics: Comparison between human intestinal biopsy sam- tory bowel disease. Nature Reviews Immunology 8(6):458–66.
ples and Colon cell lines. Drug Metabolism and Disposition doi:10.1038/nri2340.
40(4):694–705. doi:10.1124/dmd.111.042465. Collnot, E.-M., J. Susewind, and C.-M. Lehr. 2013. Advanced in vitro
Briske-Anderson, M. J., J. W. Finley, and S. M. Newman. 1997. The Models of the Intestinal Mucosa for Drug Delivery Studies. In
influence of culture time and passage number on the morphological Cellular in vitro testing: Methods and protocols, ed. J.W. Haycock, A.
and physiological development of caco-2 cells. Experimental Biology Ahluwalia, and J.M. Wilkinson, 123–133. Boca Raton: CRC Press-
and Medicine 214(3):248–57. doi:10.3181/00379727-214-44093. Taylor & Francis Group.
Brown, E. M., M. Sadarangani, and B. B. Finlay. 2013. The role of the Conklin, L. S., and M. Oliva-Hemker. 2010. Nutritional considerations
immune system in governing host-microbe interactions in the intes- in pediatric inflammatory bowel disease. Expert Review of
tine. Nature Immunology 14(7):660–7. doi:10.1038/ni.2611. Gastroenterology & Hepatology 4(3):305–17. doi:10.1586/egh.10.23.
14 
M. C. PONCE DE LEON-RODRIGUEZ ET AL.

Costantini, V., E. K. Morantz, H. Browne, K. Ettayebi, X. L. Zeng, Bischoff. 2017. ESPEN guideline: Clinical nutrition in inflammatory
R. L. Atmar, M. K. Estes, and J. Vinje. 2018. Human norovirus rep- bowel disease. Clinical Nutrition 36(2):321–47. doi:10.1016/
lication in human intestinal enteroids as model to evaluate virus j.clnu.2016.12.027.
inactivation. Emerging Infectious Diseases 24 (8):1453–64. doi: Frontela-Saseta, C., R. L opez-Nicolas, C. A. Gonzalez-Berm udez, C.
10.3201/eid2408.180126. Martınez-Gracia, and G. Ros-Berruezo. 2013. Anti-inflammatory
Denis, M.-C., Y. Desjardins, A. Furtos, V. Marcil, S. Dudonne, A. properties of fruit juices enriched with pine bark extract in an in
Montoudis, C. Garofalo, E. Delvin, A. Marette, and E. Levy. 2015. vitro model of inflamed human intestinal epithelium: The effect of
Prevention of oxidative stress, inflammation and mitochondrial dys- gastrointestinal digestion. Food and Chemical Toxicology 53 :94–9.
function in the intestine by different cranberry phenolic fractions. doi:10.1016/j.fct.2012.11.024.
Clinical Science 128(3):197–212. doi:10.1042/CS20140210. Fyderek, K., M. Strus, K. Kowalska-Duplaga, T. Gosiewski, A.
Dharmsathaphorn, K., J. A. McRoberts, K. G. Mandel, L. D. Tisdale, WeR drychowicz, U. Jedynak-Wa˛sowicz, M. Sładek, S. Pieczarkowski,
and H. Masui. 1984. A human colonic tumor cell line that maintains P. Adamski, P. Kochan, and P. B. Heczko. 2009. Mucosal bacterial
vectorial electrolyte transport. American Journal of Physiology- microflora and mucus layer thickness in adolescents with inflamma-
Gastrointestinal and Liver Physiology 246(2):G204–8. doi:10.1152/ tory bowel disease. World Journal of Gastroenterology 15(42):5287.
ajpgi.1984.246.2.G204. doi:10.3748/wjg.15.5287.
Dhuique-Mayer, C., P. Borel, E. Reboul, B. Caporiccio, P. Besancon, Galvez-Llompart, M., M. Iglesias, C. R. del, J. Galvez, and R. Garcıa-
and M.-J. Amiot. 2007. b-Cryptoxanthin From citrus juices: Domenech. 2013. Novel potential agents for ulcerative colitis by
Assessment of bioaccessibility using an in vitro digestion/caco-2 cell molecular topology: Suppression of IL-6 production in caco-2 and
culture model. British Journal of Nutrition 97(05):883–90. RAW 264.7 cell lines. Molecular Diversity 17(3):573–93. doi:10.1007/
doi:10.1017/s0007114507670822. s11030-013-9458-6.
Ding, S., and P. K. Lund. 2011. Role of intestinal inflammation as an Geremia, A., P. Biancheri, P. Allan, G. R. Corazza, and A. Di Sabatino.
early event in obesity and insulin resistance. Current Opinion in 2014. Innate and adaptive immunity in inflammatory bowel disease.
Clinical Nutrition & Metabolic Care 14(4):328–33. doi:10.1097/ Autoimmunity Reviews 13(1):3–10. doi:10.1016/j.autrev.2013.06.004.
MCO.0b013e3283478727. Giron-Calle, J., M. Alaiz, and J. Vioque. 2010. Effect of chickpea pro-
Dostal, A., M. Gagnon, C. Chassard, M. B. Zimmermann, L. tein hydrolysates on cell proliferation and in vitro bioavailability.
O’Mahony, and C. Lacroix. 2014. Salmonella adhesion, invasion and Food Research International 43(5):1365–70. doi:10.1016/
cellular immune responses are differentially affected by iron concen- j.foodres.2010.03.020.
trations in a combined in vitro gut fermentation-Cell Model. PLoS Griffith, L. G., and M. A. Swartz. 2006. Capturing complex 3D tissue
ONE 9(3):e93549. doi:10.1371/journal.pone.0093549. physiology in vitro. Nature Reviews Molecular Cell Biology
During, A., M. M. Hussain, D. W. Morel, and E. H. Harrison. 2002. 7(3):211–24. doi:10.1038/nrm1858.
Carotenoid uptake and secretion by CaCo-2 cells b-carotene isomer Hagesaether, E. 2011. Permeation modulating properties of natural pol-
selectivity and carotenoid interactions. Journal of Lipid Research ymers – effect of molecular weight and mucus. International Journal
43(7):1086–95. doi:10.1194/jlr.M200068-JLR200. of Pharmaceutics 409(1-2):150–5. doi:10.1016/j.ijpharm.2011.02.046.
Dvir-Ginzberg, M., I. Gamlieli-Bonshtein, R. Agbaria, and S. Cohen. Heller, F., Florian, P. Bojarski, C. Richter, J. Christ, M. Hillenbrand, B.
2003. Liver tissue engineering within alginate scaffolds: Effects of cell- Mankertz, J. Gitter, A. H. B€ urgel, N. Fromm. M. et al. 2005.
seeding density on hepatocyte viability, morphology, and function. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis
Tissue Engineering 9(4):757–66. doi:10.1089/107632703768247430. that affects epithelial tight junctions, apoptosis, and cell restitution.
Esch, M. B., J. H. Sung, J. Yang, C. Yu, J. Yu, J. C. March, and M. L. Gastroenterology 129(2):550–64. doi:10.1053/j.gastro.2005.05.002.
Shuler. 2012. On chip porous polymer membranes for integration of Hering, N. A., and J.-D. Schulzke. 2009. Therapeutic options to modu-
gastrointestinal tract epithelium with microfluidic ‘body-on-a-chip’ late barrier defects in inflammatory bowel disease. Digestive Diseases
devices. Biomedical Microdevices 14(5):895–906. doi:10.1007/s10544- 27(4):450–4. doi:10.1159/000233283.
012-9669-0. Hidalgo, I., T. Raub, and R. Borchardt. 1989. Characterization of the
Esser, N., S. Legrand-Poels, J. Piette, A. J. Scheen, and N. Paquot. 2014. human-Colon carcinoma cell-Line (caco-2) as a model system for
Inflammation as a link between obesity, metabolic syndrome and intestinal epithelial permeability. Gastroenterology 96(3):736–49.
type 2 diabetes. Diabetes Research and Clinical Practice doi:10.1016/0016-5085(89)90897-4.
105(2):141–50. doi:10.1016/j.diabres.2014.04.006. Hillgren, K. M., A. Kato, and R. T. Borchardt. 1995. In vitro systems
Ettayebi, K., S. E. Crawford, K. Murakami, J. R. Broughman, U. for studying intestinal drug absorption. Medicinal Research Reviews
Karandikar, V. R. Tenge, and F. H. Neill. 2016. Replication of 15(2):83–109. doi:10.1002/med.2610150202.
human noroviruses in stem cell–derived human enteroids. Science Hollebeeck, S., J. Winand, M.-F. Herent, A. During, J. Leclercq, Y.
353(6306):1387–93. doi:10.1126/science.aaf5211. Larondelle, and Y.-J. Schneider. 2012. Anti-inflammatory effects of
Ewaschuk, J. B., H. Diaz, L. Meddings, B. Diederichs, A. Dmytrash, J. pomegranate (Punica granatum L.) husk ellagitannins in caco-2
Backer, M. L. Langen, and K. L. Madsen. 2008. Secreted bioactive cells, an in vitro model of human intestine. Food & Function
factors from Bifidobacterium infantis enhance epithelial cell barrier 3(8):875–85. doi:10.1039/C2FO10258G.
function. American Journal of Physiology-Gastrointestinal and Liver Holy, C. E., M. S. Shoichet, and J. E. Davies. 2000. Engineering three-
Physiology 295(5):G1025–34. doi:10.1152/ajpgi.90227.2008. dimensional bone tissue in vitro using biodegradable scaffolds:
Fang, H.-W., S.-B. Fang, J.-S. Chiang Chiau, C.-Y. Yeung, W.-T. Chan, C.- Investigating initial cell-seeding density and culture period. Journal
B. Jiang, M.-L. Cheng, and H.-C. Lee. 2010. Inhibitory effects of of Biomedical Materials Research 51(3):376–82. doi:10.1002/1097-
Lactobacillus casei subsp. rhamnosus on salmonella lipopolysaccharide- 4636(20000905)51:3 < 376::aid-jbm11 > 3.0.co;2-g.
induced inflammation and epithelial barrier dysfunction in a co-culture Howell, S., A. Kenny, and A. Turner. 1992. A survey of membrane
model using caco-2/peripheral blood mononuclear cells. Journal of peptidases in 2 human colonic cell-Lines, caco-2 and ht-29.
Medical Microbiology 59(5):573–9. doi:10.1099/jmm.0.009662-0. Biochemical Journal 284 :595–601. doi:10.1042/bj2840595.
Fitzgerald, K. A., M. Malhotra, C. M. Curtin, F. J. O’ Brien, and C. M. Huang, C., Q. Ramadan, J. B. Wacker, H. C. Tekin, C. Ruffert, G.
O’ Driscoll. 2015. Life in 3D is never flat: 3D models to optimise Vergeres, P. Silacci, and M. A M. Gijs. 2014. Microfluidic chip for
drug delivery. Journal of Controlled Release 215 :39–54. doi:10.1016/ monitoring Ca2þ transport through a confluent layer of intestinal
j.jconrel.2015.07.020. cells. RSC Advances 4(95):52887–91. doi:10.1039/C4RA09370D.
Fogh, J., and G. Trempe. 1975. New human tumor cell lines. In Huang, H.,. J.-Q. Liu, Y. Yu, L.-H. Mo, R.-T. Ge, H.-P. Zhang, Z.-G.
Human tumor cells in vitro. ed. J. Fogh, 115–159. New York: Liu, P.-Y. Zheng, and P.-C. Yang. 2016. Regulation of TWIK-related
Springer. potassium channel-1 (Trek1) restitutes intestinal epithelial barrier
Forbes, A., J. Escher, X. Hebuterne, S. KłeR k, Z. Krznaric, S. Schneider, function. Cellular & Molecular Immunology 13(1):110–8.
R. Shamir, K. Stardelova, N. Wierdsma, A. E. Wiskin, and S. C. doi:10.1038/cmi.2014.137.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 15

Huang, S., J. M. Rutkowsky, R. G. Snodgrass, K. D. Ono-Moore, D. A. following simulated gastrointestinal digestion. Molecular Nutrition &
Schneider, J. W. Newman, S. H. Adams, and D. H. Hwang. 2012. Food Research 60(5):992–1005. doi:10.1002/mnfr.201500947.
Saturated fatty acids activate TLR-mediated proinflammatory signal- Kaulmann, A., T. Serchi, J. Renaut, L. Hoffmann, and T. Bohn. 2012.
ing pathways. Journal of Lipid Research 53(9):2002–13. doi:10.1194/ Carotenoid exposure of caco-2 intestinal epithelial cells did not
jlr.D029546. affect selected inflammatory markers but altered their proteomic
Huang, S.-M., C.-H. Wu, and G.-C. Yen. 2006. Effects of flavonoids on response. British Journal of Nutrition 108(06):963–73. doi:10.1017/
the expression of the pro-inflammatory response in human mono- S0007114511006349.
cytes induced by ligation of the receptor for AGEs. Molecular Kerneis, S., E. Caliot, H. Stubbe, A. Bogdanova, J.-P. Kraehenbuhl, and
Nutrition & Food Research 50(12):1129–39. doi:10.1002/ E. Pringault. 2000. Molecular studies of the intestinal mucosal bar-
mnfr.200600075. rier physiopathology using cocultures of epithelial and immune cells:
Huang, Z., Z. Wang, S. Long, H. Jiang, J. Chen, J. Zhang, and L. Dong. a technical update. Microbes and Infection 2(9):1119–24.
2014. A 3-D artificial Colon tissue mimic for the evaluation of doi:10.1016/S1286-4579(00)01266-1.
nanoparticle–based drug delivery system. Molecular Pharmaceutics Khor, B., A. Gardet, and R. J. Xavier. 2011. Genetics and pathogenesis
11(7):2051–61. doi:10.1021/mp400723j. of inflammatory bowel disease. Nature 474(7351):307–17.
Huet, C., C. Sahuquillo-Merino, E. Coudrier, and D. Louvard. 1987. doi:10.1038/nature10209.
Absorptive and mucus-secreting subclones isolated from a multipo- Khosravi, A., and S. K. Mazmanian. 2013. Disruption of the gut micro-
tent intestinal cell line (HT-29) provide new models for cell polarity biome as a risk factor for microbial infections. Current Opinion in
and terminal differentiation. Journal of Cell Biology 105(1):345–57. Microbiology 16(2):221–7. doi:10.1016/j.mib.2013.03.009.
doi:10.1083/jcb.105.1.345. Kim, H. J., D. Huh, G. Hamilton, and D. E. Ingber. 2012. Human gut-
Imura, Y., Y. Asano, K. Sato, and E. Yoshimura. 2009. A microfluidic on-a-chip inhabited by microbial flora that experiences intestinal
system to evaluate intestinal absorption. Analytical Sciences peristalsis-like motions and flow. Lab on a Chip 12(12):2165–74.
25(12):1403–7. doi:10.2116/analsci.25.1403. doi:10.1039/C2LC40074J.
Jaffe, A. B., N. Kaji, J. Durgan, and A. Hall. 2008. Cdc42 controls spin- Kim, H. J., and D. E. Ingber. 2013. Gut-on-a-Chip Microenvironment
dle orientation to position the apical surface during epithelial mor- induces human intestinal cells to undergo villus differentiation.
phogenesis. The Journal of Cell Biology 183(4):625–33. doi:10.1083/ Integrative Biology 5(9):1130–40. doi:10.1039/C3IB40126J.
jcb.200807121. Kim, K.-M., Y.-S. Kim, J. Y. Lim, S. J. Min, H.-C. Ko, S.-J. Kim, and Y.
Jiang, H. A. I.-Y. U. E., F. E. N. G. Wang, H. A. I.-M. I. N. Chen, and Kim. 2015. Intestinal anti-inflammatory activity of sasa quelpaerten-
X. I. A. O.-J. U. N. Yan. 2013. j-carrageenan induces the disruption sis leaf extract by suppressing lipopolysaccharide-stimulated inflam-
of intestinal epithelial caco-2 monolayers by promoting the inter- matory mediators in intestinal epithelial caco-2 cells co-cultured
action between intestinal epithelial cells and immune cells. with RAW 264.7 macrophage cells. Nutrition Research and Practice
Molecular Medicine Reports 8(6):1635–42. doi:10.3892/ 9(1):3. doi:10.4162/nrp.2015.9.1.3.
mmr.2013.1726. Kim, S. H., M. Chi, B. Yi, S. H. Kim, S. Oh, Y. Kim, S. Park, and J. H.
Juuti-Uusitalo, K., L. J. Klunder, K. A. Sjollema, K. Mackovicova, R. Sung. 2014. Three-dimensional intestinal villi epithelium enhances
Ohgaki, D. Hoekstra, J. Dekker, and S. C. D. van IJzendoorn. 2011. protection of human intestinal cells from bacterial infection by
Differential effects of TNF (TNFSF2) and IFN-c on intestinal epithe- inducing mucin expression. Integrative Biology 6(12):1122–31.
lial cell morphogenesis and barrier function in three-Dimensional doi:10.1039/C4IB00157E.
Culture. PLoS ONE 6(8):e22967. doi:10.1371/journal.pone.0022967. Kimura, H., T. Yamamoto, H. Sakai, Y. Sakai, and T. Fujii. 2008. An
Juuti-Uusitalo, K. M., K. Kaukinen, M. M€aki, J. Tuimala, and H. integrated microfluidic system for long-term perfusion culture and
Kainulainen. 2006. Gene expression in TGFbeta-induced epithelial on-line monitoring of intestinal tissue models. Lab on a Chip
cell differentiation in a three-dimensional intestinal epithelial cell 8(5):741–6. doi:10.1039/B717091B.
differentiation model. BMC Genomics 7 :279. doi:10.1186/1471-2164- de Kivit, S., E. van Hoffen, N. Korthagen, J. Garssen, and L. E. M.
7-279. Willemsen. 2011. Apical TLR ligation of intestinal epithelial cells
Kamada, N., and G. N ~ez. 2014. Regulation of the immune system by
un drives a Th1-polarized regulatory or inflammatory type effector
the resident intestinal bacteria. Gastroenterology 146(6):1477–88. response in vitro. Immunobiology 216(4):518–27. doi:10.1016/
doi:10.1053/j.gastro.2014.01.060. j.imbio.2010.08.005.
Kamada, N., S.-U. Seo, G. Y. Chen, and G. N ~ez. 2013. Role of the
un Kleiveland, C. R. 2015. Peripheral blood mononuclear cells. In The
gut microbiota in immunity and inflammatory disease. Nature impact of food bioactives on health. eds. K. Verhoeckx, P. Cotter, I.
Reviews Immunology 13(5):321–35. doi:10.1038/nri3430. Lopez-Exposito, C. Kleiveland, T. Lea, A. Mackie, T. Requena, D.
Kang, K.-H., C.-S. Kong, Y. Seo, M.-M. Kim, and S.-K. Kim. 2009. Swiatecka, and H. Wichers, 161–167. New York, NY: Springer
Anti-inflammatory effect of coumarins isolated from corydalis heter- International Publishing.
ocarpa in HT-29 human Colon carcinoma cells. Food and Chemical Kosi nska, A., and W. Andlauer. 2012. Cocoa polyphenols are absorbed
Toxicology 47(8):2129–34. doi:10.1016/j.fct.2009.05.036. in caco-2 cell model of intestinal epithelium. Food Chemistry
Kanzato, H., M. Manabe, and M. Shimizu. 2001. An in vitro approach 135(3):999–1005. doi:10.1016/j.foodchem.2012.05.101.
to the evaluation of the cross talk between intestinal epithelium and Kosi nska, A., and W. Andlauer. 2013. Modulation of tight junction
macrophages. Bioscience, Biotechnology, and Biochemistry integrity by food components. Food Research International
65(2):449–51. doi:10.1271/bbb.65.449. 54(1):951–60. doi:10.1016/j.foodres.2012.12.038.
Kasper, J. Y., M. I. Hermanns, C. Cavelius, A. Kraegeloh, T. Jung, R. Kostic, A. D., R. J. Xavier, and D. Gevers. 2014. The microbiome in
Danzebrink, R. E. Unger, and C. J. Kirkpatrick. 2016. The role of inflammatory bowel disease: Current status and the future ahead.
the intestinal microvasculature in inflammatory bowel disease: Gastroenterology 146(6):1489–99. doi:10.1053/j.gastro.2014.02.009.
Studies with a modified caco-2 model including endothelial cells Kuan, S.-F., J. C. Byrd, C. B. Basbaum, and Y. S. Kim. 1987.
resembling the intestinal barrier in vitro. International Journal of Characterization of quantitative mucin variants from a human
Nanomedicine Volume 11 :6353–64. doi:10.2147/IJN.S92608. Colon cancer cell line. Cancer Research 47(21):5715–24.
Kaulmann, A., C. M. Andre, Y.-J. Schneider, L. Hoffmann, and T. Langerholc, T., P. A. Maragkoudakis, J. Wollgast, L. Gradisnik, and A.
Bohn. 2016. Carotenoid and polyphenol bioaccessibility and cellular Cencic. 2011. Novel and established intestinal cell line models – An
uptake from plum and cabbage varieties. Food Chemistry em 197 indispensable tool in food science and nutrition. Trends in Food
Part A: :325–32. doi:10.1016/j.foodchem.2015.10.049. Science & Technology 22 :S11–S20. doi:10.1016/j.tifs.2011.03.010.
Kaulmann, A., S. Legay, Y.-J. Schneider, L. Hoffmann, and T. Bohn. Langhans, S. A. 2018. Three-Dimensional In vitro cell culture models
2016. Inflammation related responses of intestinal cells to plum and in drug discovery and drug repositioning. Frontiers in Pharmacology
cabbage digesta with differential carotenoid and polyphenol profiles 9:6. doi:10.3389/fphar.2018.00006.
16 
M. C. PONCE DE LEON-RODRIGUEZ ET AL.

Laparra, J. M., R. P. Glahn, and D. D. Miller. 2009. Different responses Nutritional Biochemistry 20(7):494–502. doi:10.1016/
of Fe transporters in caco-2/HT29-MTX cocultures than in inde- j.jnutbio.2008.05.006.
pendent caco-2 cell cultures. Cell Biology International 33(9):971–7. Makon-Sebastien, N., F. Francis, S. Eric, V. P. Henri, L. J. François, P.
doi:10.1016/j.cellbi.2009.06.001. Laurent, B. Yves, and C. Serge. 2014. Lycopene modulates THP1
Lea, T. 2015. Caco-2 Cell Line. In The impact of food bioactives on and Caco2 cells inflammatory state through transcriptional and non-
health., ed. K. Verhoeckx, P. Cotter, I. L opez-Exp
osito, C. transcriptional processes. Mediators of Inflammation 2014 :1.
Kleiveland, T. Lea, A. Mackie, T. Requena, D. Swiatecka, and H. doi:10.1155/2014/507272, 10.1155/2014/507272.
Wichers, 103–111. New York, NY: Springer International Mallon, D. P., and D. L. Suskind. 2010. Nutrition in pediatric inflam-
Publishing. matory bowel disease. Nutrition in Clinical Practice 25(4):335–9.
Lefebvre, D. E., K. Venema, L. Gombau, L. G. Valerio, J. Raju, G. S. doi:10.1177/0884533610373773.
Bondy, H. Bouwmeester, R. P. Singh, A. J. Clippinger, E.-M. Manichanh, C., N. Borruel, F. Casellas, and F. Guarner. 2012. The gut
Collnot, et al., and V. Stone. 2015. Utility of models of the gastro- microbiota in IBD. Nature Reviews Gastroenterology & Hepatology
intestinal tract for assessment of the digestion and absorption of 9(10):599–608. doi:10.1038/nrgastro.2012.152.
engineered nanomaterials released from food matrices. Martins, I. M., G. A. Macedo, J. A. Macedo, B. S. Roberto, Q. Chen,
Nanotoxicology 9(4):523–42. doi:10.3109/17435390.2014.948091. J. B. Blumberg, and C.-Y. O. Chen. 2017. Tannase enhances the
Leibovitz, A., J. C. Stinson, W. B. McCombs, C. E. McCoy, K. C. anti-inflammatory effect of grape pomace in caco-2 cells treated
Mazur, and N. D. Mabry. 1976. Classification of human colorectal with IL-1b. Journal of Functional Foods 29 :69–76. doi:10.1016/
adenocarcinoma cell lines. Cancer Research 36(12):4562–9. j.jff.2016.12.011.
Leonard, F., H. Ali, E.-M. Collnot, B. J. Crielaard, T. Lammers, G. Maslowski, K. M., Vieira, A. T. Ng, A. Kranich, J. Sierro, F. Yu, D.
Storm, and C.-M. Lehr. 2012. Screening of budesonide nanoformula- Schilter, H. C. Rolph, M. S. Mackay, F. Artis. D., et al. 2009.
tions for treatment of inflammatory bowel disease in an inflamed Regulation of inflammatory responses by gut microbiota and
3D Cell-Culture Model. ALTEX: Alternatives to Animal Experiments chemoattractant receptor GPR43. Nature 461(7268):1282–6.
29(3):275–85. doi:10.14573/altex.2012.3.275. doi:10.1038/nature08530.
Leonard, F., E.-M. Collnot, and C.-M. Lehr. 2010. A three-dimensional Massironi, S., R. E. Rossi, F. A. Cavalcoli, S. Della Valle, M. Fraquelli,
coculture of enterocytes, monocytes and dendritic cells to model and D. Conte. 2013. Nutritional deficiencies in inflammatory bowel
inflamed intestinal mucosa in vitro. Molecular Pharmaceutics disease: Therapeutic approaches. Clinical Nutrition 32(6):904–10.
7(6):2103–19. doi:10.1021/mp1000795. doi:10.1016/j.clnu.2013.03.020.
Lesuffleur, T., A. Barbat, E. Dussaulx, and A. Zweibaum. 1990. Growth Matias, A., S. L. Nunes, J. Poejo, E. Mecha, A. T. Serra, P. J. A.
adaptation to methotrexate of HT-29 human colon carcinoma cells Madeira, M. R. Bronze, and C. M. M. Duarte. 2014. Antioxidant
is associated with their ability to differentiate into columnar absorp- and anti-inflammatory activity of a flavonoid-rich concentrate recov-
tive and mucus-secreting cells. Cancer Research 50(19):6334–43. ered from Opuntia ficus-indica juice. Food & Function
Lesuffleur, T., N. Porchet, J. P. Aubert, D. Swallow, J. R. Gum, Y. S. 5(12):3269–80. doi:10.1039/C4FO00071D.
Kim, F. X. Real, and A. Zweibaum. 1993. Differential expression of Matsusaki, M.,. D. Hikimoto, A. Nishiguchi, K. Kadowaki, K. Ohura,
the human mucin genes MUC1 to MUC5 in relation to growth and T. Imai, and M. Akashi. 2015. 3D-fibroblast tissues constructed by a
differentiation of different mucus-secreting HT-29 cell subpopula- cell-coat technology enhance tight-junction formation of human
tions. Journal of Cell Science 106(3):771–83. colon epithelial cells. Biochemical and Biophysical Research
Li, N., D. Wang, Z. Sui, X. Qi, L. Ji, X. Wang, and L. Yang. 2013. Communications 457(3):363–9. doi:10.1016/j.bbrc.2014.12.118.
Development of an improved three-Dimensional In vitro intestinal McCracken, K. W., J. C. Howell, J. M. Wells, and J. R. Spence. 2011.
mucosa model for drug absorption evaluation. Tissue Eng. Part C Generating human intestinal tissue from pluripotent stem cells in
Methods 19(9):708–19. doi:10.1089/ten.tec.2012.0463. vitro. Nature Protocols 6 (12):1920–8. doi:10.1038/nprot.2011.410.
Liang, G. H., and C. R. Weber. 2014. Molecular aspects of tight junc- Middendorp, S., K. Schneeberger, C. L. Wiegerinck, M. Mokry,
tion barrier function. Current Opinion in Pharmacology 19 :84–9. R. D. L. Akkerman, S. van Wijngaarden, H. Clevers, and E. E. S.
doi:10.1016/j.coph.2014.07.017. Nieuwenhuis. 2014. Adult stem cells in the small intestine are
Liu, T.-C., and T. S. Stappenbeck. 2016. Genetics and pathogenesis of intrinsically programmed with their location-Specific Function. Stem
inflammatory bowel disease. Annual Review of Pathology: Cells 32 (5):1083–91. doi:10.1002/stem.1655.
Mechanisms of Disease 11(1):127–48. doi:10.1146/annurev-pathol- Mijac, D. D., G. L. J. Jankovic, J. Jorga, and M. N. Krstic. 2010.
012615-044152. Nutritional status in patients with active inflammatory bowel dis-
Loftus, E. V. 2004. Clinical epidemiology of inflammatory bowel dis- ease: Prevalence of malnutrition and methods for routine nutritional
ease: Incidence, prevalence, and environmental influences. assessment. European Journal of Internal Medicine 21(4):315–9.
Gastroenterology 126(6):1504–17. doi:10.1053/j.gastro.2004.01.063. doi:10.1016/j.ejim.2010.04.012.
Maccaferri, S., A. Klinder, P. Brigidi, P. Cavina, and A. Costabile. 2012. Molodecky, N. A., I. S. Soon, D. M. Rabi, W. A. Ghali, M. Ferris, G.
Potential probiotic Kluyveromyces marxianus B0399 modulates the Chernoff, E. I. Benchimol, R. Panaccione, S. Ghosh, H. W. Barkema,
immune response in caco-2 cells and peripheral blood mononuclear and G. G. Kaplan. 2012. Increasing incidence and prevalence of the
cells and impacts the human gut microbiota in an in vitro colonic inflammatory bowel diseases with time, based on systematic review.
model system. Applied and Environmental Microbiology Gastroenterology 142(1):46–54.e42. doi:10.1053/j.gastro.2011.10.001.
78(4):956–64. doi:10.1128/AEM.06385-11. Mori, A., H. Satsu, and M. Shimizu. 2003. New model for studying the
Madara, J. L., and K. Dharmsathaphorn. 1985. Occluding junction migration of immune cells into intestinal epithelial cell monolayers.
structure-function relationships in a cultured epithelial monolayer. Cytotechnology 43(1-3):57–64. doi:10.1023/B:
Journal of Cell Biology 101(6):2124–33. doi:10.1083/jcb.101.6.2124. CYTO.0000039910.30540.8f.
Maher, S., R. J. Mrsny, and D. J. Brayden. 2016. Intestinal permeation Moyes, S. M., J. F. Morris, and K. E. Carr. 2010. Macrophages increase
enhancers for oral peptide delivery. Advanced Drug Delivery Reviews microparticle uptake by enterocyte-like caco-2 cell monolayers.
106PartB:277–319. doi:10.1016/j.addr.2016.06.005. Journal of Anatomy 217(6):740–54. doi:10.1111/j.1469-
Mahler, G. J., M. B. Esch, R. P. Glahn, and M. L. Shuler. 2009. 7580.2010.01304.x.
Characterization of a gastrointestinal tract microscale cell culture Murakami, H., and H. Masui. 1980. Hormonal control of human colon
analog used to predict drug toxicity. Biotechnology and carcinoma cell growth in serum-free medium. Proceedings of the
Bioengineering 104(1):193–205. doi:10.1002/bit.22366. National Academy of Sciences 77(6):3464–8. doi:10.1073/
Mahler, G. J., M. L. Shuler, and R. P. Glahn. 2009. Characterization of pnas.77.6.3464.
caco-2 and HT29-MTX cocultures in an in vitro digestion/cell cul- Nerurkar, M. M., P. S. Burton, and R. T. Borchardt. 1996. The use of
ture model used to predict iron bioavailability. The Journal of surfactants to enhance the permeability of peptides through caco-2
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 17

cells by inhibition of an apically polarized efflux system. Gijs. 2013. NutriChip: Nutrition analysis meets microfluidics. Lab
Pharmaceutical Research 13(4):528–34. doi:10.1023/A:1016033702220. on a Chip 13(2):196–203. doi:10.1039/C2LC40845G.
Neurath, M. F. 2014. Cytokines in inflammatory bowel disease. Nature Ramadan, Q., and L. Jing. 2016. Characterization of tight junction dis-
Reviews Immunology 14(5):329–42. doi:10.1038/nri3661. ruption and immune response modulation in a miniaturized caco-2/
Ng, S. C., C. N. Bernstein, M. H. Vatn, P. L. Lakatos, E. V. Loftus, C. U937 coculture-based in vitro model of the human intestinal barrier.
Tysk, C. O’Morain, B. Moum, and J.-F. Colombel. and (ioibd), on Biomedical Microdevices 18(1):11. doi:10.1007/s10544-016-0035-5.
behalf of the E. and N.H.T.F. of the I.O. of I.B.D. 2013. Geographical Ranaldi, G., R. Consalvo, Y. Sambuy, and M. L. Scarino. 2003.
variability and environmental risk factors in inflammatory bowel dis- Permeability characteristics of parental and clonal human intestinal
ease. Gut 62(4):630–49. doi:10.1136/gutjnl-2012-303661. caco-2 cell lines differentiated in serum-supplemented and serum-
Nichols, J. M., I. Maiellaro, J. Abi-Jaoude, S. Curci, and A. M. Hofer. free media. Toxicology in Vitro 17(5-6):761–7. doi:10.1016/S0887-
2015. Store-operated” cAMP signaling contributes to Ca2þ-activated 2333(03)00095-X.
cl  secretion in T84 colonic cells. American Journal of Physiology- Raschke, W. C., S. Baird, P. Ralph, and I. Nakoinz. 1978. Functional
Gastrointestinal and Liver Physiology 309(8):G670–9. doi:10.1152/ macrophage cell lines transformed by abelson leukemia virus. Cell
ajpgi.00214.2015. 15(1):261–7. doi:10.1016/0092-8674(78)90101-0.
Nollevaux, G., C. Deville, B. El Moualij, W. Zorzi, P. Deloyer, Y.-J. Ravi, M., V. Paramesh, S. R Kaviya, E. Anuradha, and F. D. P.
Schneider, O. Peulen, and G. Dandrifosse. 2006. Development of a Solomon. 2015. 3D cell culture systems: Advantages and applica-
serum-free co-culture of human intestinal epithelium cell-lines tions. Journal of Cellular Physiology 230(1):16–26. doi:10.1002/
(caco-2/HT29-5M21). BMC Cell Biology 7(1):20. doi:10.1186/1471- jcp.24683.
2121-7-20. Reiff, C., and D. Kelly. 2010. Inflammatory bowel disease, gut bacteria
Nusrat, A., C. V Eichel-Streiber, J. R. Turner, P. Verkade, J. L. Madara, and probiotic therapy. International Journal of Medical Microbiology
and C. A. Parkos. 2001. Clostridium difficile toxins disrupt epithelial 300(1):25–33. doi:10.1016/j.ijmm.2009.08.004.
barrier function by altering membrane microdomain localization of Des Rieux, A., V. Fievez, I. Theate, J. Mast, V. Preat, and Y.-J.
tight junction proteins. Infection and Immunity 69(3):1329–36. Schneider. 2007. An improved in vitro model of human intestinal
doi:10.1128/IAI.69.3.1329-1336.2001. follicle-associated epithelium to study nanoparticle transport by M
O’Hara, J. R., and A. G. Buret. 2008. Mechanisms of intestinal tight cells. European Journal of Pharmaceutical Sciences 30(5):380–91.
junctional disruption during infection. Frontiers in Bioscience 13 doi:10.1016/j.ejps.2006.12.006.
:7008–21. doi:10.2741/3206. Rodrıguez-Ramiro, I., S. Ramos, E. Lopez-Oliva, A. Agis-Torres, L.
Ohnuma, T., H. Arkin, and J. F. Holland. 1986. Effects of cell density Bravo, L. Goya, and M. A. Martın. 2013. Cocoa polyphenols prevent
on drug-induced cell kill kinetics in vitro (inoculum effect). British inflammation in the Colon of azoxymethane-treated rats and in
Journal of Cancer 54(3):415–21. doi:10.1038/bjc.1986.192. TNF-a-stimulated caco-2 cells. British Journal of Nutrition
Olejnik, A., K. Kowalska, M. Olkowicz, J. Rychlik, W. Juzwa, K. 110(02):206–15. doi:10.1017/S0007114512004862.
Myszka, R. Dembczy nski, and W. Białas. 2015. Anti-inflammatory Ryan, S.-L., A.-M. Baird, G. Vaz, A. J. Urquhart, h Senge, D. J. Richard,
effects of gastrointestinal digested Sambucus nigra L. fruit extract K. J. O’Byrne, and A. M. Davies. 2016. Drug discovery approaches uti-
analysed in co-cultured intestinal epithelial cells and lipopolysac- lizing three-dimensional cell culture. ASSAY and Drug Development
charide-stimulated macrophages. Journal of Functional Foods 19 Technologies 14(1):19–28. doi:10.1089/adt.2015.670.
:649–60. Part A: doi:10.1016/j.jff.2015.09.064. Salerno-Goncalves, R., A. Fasano, and M. B. Sztein. 2011. Engineering of
Otte, J.-M., and D. K. Podolsky. 2004. Functional modulation of enter- a multi-cellular organotypic model of the human intestinal mucosa.
ocytes by gram-positive and gram-negative microorganisms. Gastroenterology 141(2):e18–20. doi:10.1053/j.gastro.2011.04.062.
American Journal of Physiology-Gastrointestinal and Liver Physiology Samak, G., S. Aggarwal, and R. K. Rao. 2011. ERK is involved in EGF-
286(4):G613–26. doi:10.1152/ajpgi.00341.2003. mediated protection of tight junctions, but not adherens junctions,
Ou, G., V. Baranov, E. Lundmark, S. Hammarstr€om, and M.-L. in acetaldehyde-treated caco-2 cell monolayers. American Journal of
Hammarstr€ om. 2009. Contribution of intestinal epithelial cells to Physiology-Gastrointestinal and Liver Physiology 301(1):G50–9.
innate immunity of the human gut – Studies on polarized mono- doi:10.1152/ajpgi.00494.2010.
layers of Colon carcinoma cells. Scandinavian Journal of Sambuy, Y., I. D. Angelis, G. Ranaldi, M. L. Scarino, A. Stammati, and
Immunology 69(2):150–61. doi:10.1111/j.1365-3083.2008.02208.x. F. Zucco. 2005. The caco-2 cell line as a model of the intestinal bar-
Pampaloni, F., E. G. Reynaud, and E. H. K. Stelzer. 2007. The third rier: influence of cell and culture-related factors on caco-2 cell func-
dimension bridges the gap between cell culture and live tissue. tional characteristics. Cell Biology and Toxicology 21(1):1–26.
Nature Reviews Molecular Cell Biology 8(10):839–45. doi:10.1038/ doi:10.1007/s10565-005-0085-6.
nrm2236. Sato, T., D. E. Stange, M. Ferrante, R. G. J. Vries, J. H. van Es, S. van
Pandolfi, F., R. Cianci, D. Pagliari, R. Landolfi, and G. Cammarota. den Brink, W. J. van Houdt, A. Pronk, J. van Gorp, P. D. Siersema,
2009. Cellular mediators of inflammation: Tregs and TH cells in and H. Clevers. 2011. Long-Term Expansion of epithelial organoids
gastrointestinal diseases. Mediators Inflamm 2009 :1. doi:10.1155/ from human colon, adenoma, adenocarcinoma, and Barrett’s epithe-
2009/132028. lium. Gastroenterology 141 (5):1762–72. doi:10.1053/
Parlesak, A., D. Haller, S. Brinz, A. Baeuerlein, and C. Bode. 2004. j.gastro.2011.07.050.
Modulation of cytokine release by differentiated CACO-2 cells in a Satsu, H., Y. Ishimoto, T. Nakano, T. Mochizuki, T. Iwanaga, and M.
compartmentalized coculture model with mononuclear leucocytes Shimizu. 2006. Induction by activated macrophage-like THP-1 cells
and nonpathogenic bacteria. Scandinavian Journal of Immunology of apoptotic and necrotic cell death in intestinal epithelial caco-2
60(5):477–85. doi:10.1111/j.0300-9475.2004.01495.x. monolayers via tumor necrosis factor-alpha. Experimental Cell
Pinto, M., S. Robineleon, M. Appay, M. Kedinger, N. Triadou, E. Research 312(19):3909–19. doi:10.1016/j.yexcr.2006.08.018.
Dussaulx, B. Lacroix, P. Simonassmann, K. Haffen, J. Fogh, and A. Saxena, K., S. E. Blutt, K. Ettayebi, X.-L. Zeng, R. B. James, E. C. Sue,
Zweibaum. 1983. Enterocyte-like differentiation and polarization of and U. Karandikar. 2015. Human intestinal enteroids: A new model
the human-Colon carcinoma cell-Line Caco-2 in culture. Biology of to study human rotavirus infection, host restriction and pathophysi-
the Cell 47(3):323–30. ology. Journal of Virology 90(1):43–56. doi:10.1128/JVI.01930-15.
Poquet, L., M. N. Clifford, and G. Williamson. 2008. Transport and Saxena, K., L. M. Simon, X.-L. Zeng, S. E. Blutt, S. E. Crawford, N. P.
metabolism of ferulic acid through the colonic epithelium. Drug Sastri, and U. C. Karandikar. 2017. A paradox of transcriptional and
Metabolism and Disposition 36(1):190–7. doi:10.1124/ functional innate interferon responses of human intestinal enteroids
dmd.107.017558. to enteric virus infection. Proceedings of the National Academy of
Ramadan, Q., H. Jafarpoorchekab, C. Huang, P. Silacci, S. Carrara, G. Sciences of the United States of America 114(4):E570–79.
Kokl€ u, J. Ghaye, J. Ramsden, C. Ruffert, G. Vergeres, and M. A. M. doi:10.1073/pnas.1615422114.
18 
M. C. PONCE DE LEON-RODRIGUEZ ET AL.

Schimpel, C., B. Teubl, M. Absenger, C. Meindl, E. Fr€ ohlich, G. Tanoue, T.,. Y. Nishitani, K. Kanazawa, T. Hashimoto, and M. Mizuno.
Leitinger, A. Zimmer, and E. Roblegg. 2014. Development of an 2008. In vitro model to estimate gut inflammation using co-cultured
advanced intestinal in vitro triple culture permeability model to caco-2 and RAW264.7 cells. Biochemical and Biophysical Research
study transport of nanoparticles. Molecular Pharmaceutics Communications 374(3):565–9. doi:10.1016/j.bbrc.2008.07.063.
11(3):808–18. doi:10.1021/mp400507g. Tom, B. H., L. P. Rutzky, M. M. Jakstys, R. Oyasu, C. I. Kaye, and
Selby-Pham, S. N. B., S. A. Osborne, K. S. Howell, F. R. Dunshea, and B. D. Kahan. 1976. Human colonic adenocarcinoma cells. In Vitro
L. E. Bennett. 2017. Transport rates of dietary phytochemicals in cell 12(3):180–91. doi:10.1007/BF02796440.
monolayers is inversely correlated with absorption kinetics in Tsuchiya, S., M. Yamabe, Y. Yamaguchi, Y. Kobayashi, T. Konno, and
humans. Journal of Functional Foods 39 :206–14. doi:10.1016/ K. Tada. 1980. Establishment and characterization of a human acute
j.jff.2017.10.016. monocytic leukemia cell line (THP-1). International Journal of
Sergent, T., N. Piront, J. Meurice, O. Toussaint, and Y.-J. Schneider. Cancer 26(2):171–6. doi:10.1002/ijc.2910260208.
2010. Anti-inflammatory effects of dietary phenolic compounds in Turco, L., T. Catone, F. Caloni, E. D. Consiglio, E. Testai, and A.
an in vitro model of inflamed human intestinal epithelium. Stammati. 2011. Caco-2/TC7 cell line characterization for intestinal
Chemico-Biological Interactions 188(3):659–67. doi:10.1016/ absorption: How reliable is this in vitro model for the prediction of
j.cbi.2010.08.007. the oral dose fraction absorbed in human? Toxicology in Vitro
Shanahan, F., and E. M. M. Quigley. 2014. Manipulation of the micro- 25(1):13–20. doi:10.1016/j.tiv.2010.08.009.
biota for treatment of IBS and IBD—challenges and controversies. Turner, J. R. 2006. Molecular basis of epithelial barrier regulation:
Gastroenterology 146(6):1554–63. doi:10.1053/j.gastro.2014.01.050. From basic mechanisms to clinical application. The American
Sheehan, D., C. Moran, and F. Shanahan. 2015. The microbiota in Journal of Pathology 169(6):1901–9. doi:10.2353/ajpath.2006.060681.
inflammatory bowel disease. Journal of Gastroenterology Turner, J. R. 2009. Intestinal mucosal barrier function in health and
50(5):495–507. doi:10.1007/s00535-015-1064-1. disease. Nature Reviews Immunology 9(11):799–809. doi:10.1038/
Shim, K.-Y., D. Lee, J. Han, N.-T. Nguyen, S. Park, and J. H. Sung. nri2653.
2017. Microfluidic gut-on-a-chip with three-dimensional villi struc- Turpin, W., C. Humblot, M.-L. Noordine, M. Thomas, and J.-P.
ture. Biomedical Microdevices 19(2):37. doi:10.1007/s10544-017-0179- Guyot. 2012. Lactobacillaceae and cell adhesion: Genomic and
y. functional screening. PLoS ONE 7(5):e38034. doi:10.1371/
Shimizu, M. 2010. Interaction between food substances and the intes- journal.pone.0038034.
tinal epithelium. Bioscience, Biotechnology, and Biochemistry Tyrer, P., A. Ruth Foxwell, J. Kyd, M. Harvey, P. Sizer, and A. Cripps.
74(2):232–41. doi:10.1271/bbb.90730. 2002. Validation and quantitation of an in vitro M-cell model.
Shimizu, M. 2017. Multifunctions of dietary polyphenols in the regula- Biochemical and Biophysical Research Communications
tion of intestinal inflammation. Journal of Food and Drug Analysis
299(3):377–83. doi:10.1016/S0006-291X(02)02631-1.
25(1):93–9. doi:10.1016/j.jfda.2016.12.003. Tyrer, P. C., E. G. Bean, A. Ruth Foxwell, and P. Pavli. 2011. Effects of
Simonovic, I., J. Rosenberg, A. Koutsouris, and G. Hecht. 2000.
bacterial products on enterocyte–macrophage interactions in vitro.
Enteropathogenic Escherichia coli dephosphorylates and dissociates
Biochemical and Biophysical Research Communications
occludin from intestinal epithelial tight junctions. Cellular
413(2):336–41. doi:10.1016/j.bbrc.2011.08.100.
Microbiology 2(4):305–15. doi:10.1046/j.1462-5822.2000.00055.x.
Utech, M., A. I. Ivanov, S. N. Samarin, M. Bruewer, J. R. Turner, R. J.
Smith, P. M., M. R. Howitt, N. Panikov, M. Michaud, C. A. Gallini, M.
Mrsny, C. A. Parkos, and A. Nusrat. 2005. Mechanism of IFN-
Bohlooly-Y, J. N. Glickman, and W. S. Garrett. 2013. The microbial
c-induced endocytosis of tight junction proteins: Myosin II-depend-
metabolites, short-Chain Fatty acids, regulate colonic treg cell
ent vacuolarization of the apical plasma membrane. Molecular
homeostasis. Science 341(6145):569–73. doi:10.1126/science.1241165.
Biology of the Cell 16(10):5040–52. doi:10.1091/mbc.E05-03-0193.
Spence, J. R., C. N. Mayhew, S. A. Rankin, M. F. Kuhar, J. E. Vallance,
Vachon, P., and J. Beaulieu. 1992. Transient mosaic patterns of mor-
K. Tolle, E. E. Hoskins, V. V. Kalinichenko, S. I. Wells, A. M. Zorn,
et al. 2011. Directed differentiation of human pluripotent stem cells phological and functional-Differentiation In the caco-2 cell-Line.
into intestinal tissue in vitro. Nature 470 (7332):105–9. doi:10.1038/ Gastroenterology 103(2):414–23. doi:10.1016/0016-5085(92)90829-n.
nature09691. Vagianos, K., S. Bector, J. McConnell, and C. N. Bernstein. 2007.
Strober, W., I. J. Fuss, and R. S. Blumberg. 2002. The immunology of Nutrition assessment of patients with inflammatory bowel disease.
mucosal models of inflammation. Annual Review of Immunology Journal of Parenteral and Enteral Nutrition 31(4):311–9. doi:10.1177/
20(1):495–549. doi:10.1146/annurev.immunol.20.100301.064816. 0148607107031004311.
Susewind, J., C. Carvalho-Wodarz, S. de, U. Repnik, E.-M. Collnot, N. Vamadevan, A. S., M. Fukata, E. T. Arnold, L. S. Thomas, D. Hsu, and
Schneider-Daum, G. W. Griffiths, and C.-M. Lehr. 2016. A 3D co- M. T. Abreu. 2010. Regulation of toll-like receptor 4-associated MD-
culture of three human cell lines to model the inflamed intestinal 2 in intestinal epithelial cells: a comprehensive analysis. Innate
mucosa for safety testing of nanomaterials. Nanotoxicology Immunity 16(2):93–103. doi:10.1177/1753425909339231.
10(1):1–62. doi:10.3109/17435390.2015.1008065. VanDussen, K. L., J. M. Marinshaw, N. Shaikh, H. Miyoshi, C. Moon,
Suzuki, M., T. Hisamatsu, and D. K. Podolsky. 2003. Gamma inter- P. I. Tarr, M. A. Ciorba, and T. S. Stappenbeck. 2014. Development
feron augments the intracellular pathway for lipopolysaccharide of an enhanced human gastrointestinal epithelial culture system to
(LPS) recognition in human intestinal epithelial cells through coor- facilitate patient-based assays. Gut 64(6):911–20. doi:10.1136/gutjnl-
dinated up-regulation of LPS uptake and expression of the intracel- 2013-306651.
lular toll-like receptor 4-MD-2 complex. Infection and Immunity Vergeres, G., Bogicevic, B. Buri, C. Carrara, S. Chollet, M. Corbino-
71(6):3503–11. doi:10.1128/IAI.71.6.3503-3511.2003. Giunta, L. Egger, L. Gille, D. Kopf-Bolanz, K. Laederach. K. et al.
Suzuki, T. 2013. Regulation of intestinal epithelial permeability by tight 2012. The NutriChip project – translating technology into nutri-
junctions. Cellular and Molecular Life Sciences 70(4):631–59. tional knowledge. British Journal of Nutrition 108(05):762–8.
doi:10.1007/s00018-012-1070-x. doi:10.1017/S0007114512002693.
Suzuki, T., S. Tanabe, and H. Hara. 2011. Kaempferol enhances intes- Viallard, V.,. C. Denis, V. Trocheris, and J. C. Murat. 1986. Effect of
tinal barrier function through the cytoskeletal association and glutamine deprivation and glutamate or ammonium chloride add-
expression of tight junction proteins in caco-2 cells. The Journal of ition on growth rate, metabolism and differentiation of human
Nutrition 141(1):87–94. doi:10.3945/jn.110.125633. Colon cancer cell-line HT29. International Journal of Biochemistry
Tang, J., P. Bouyer, A. Mykoniatis, M. Buschmann, K. S. Matlin, and 18(3):263–9. doi:10.1016/0020-711X(86)90116-3.
J. B. Matthews. 2010. Activated PKCd and PKC inhibit epithelial Vieira, E. L. M., A. J. Leonel, A. P. Sad, N. R. M. Beltr~ao, T. F. Costa,
chloride secretion response to cAMP via inducing internalization of T. M. R. Ferreira, A. C. Gomes-Santos, A. M. C. Faria, M. C. G.
the naþ-Kþ-2Cl  cotransporter NKCC1. Journal of Biological Peluzio, D. C. Cara, and J. I. Alvarez-Leite. 2012. Oral administra-
Chemistry 285(44):34072–85. doi:10.1074/jbc.M110.137380. tion of sodium butyrate attenuates inflammation and mucosal lesion
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 19

in experimental acute ulcerative colitis. International Journal of Wu, G. D., Chen, J. Hoffmann, C. Bittinger, K. Chen, Y.-Y. Keilbaugh,
Biochemistry 23(5):430–6. doi:10.1016/j.jnutbio.2011.01.007. S. A. Bewtra, M. Knights, D. Walters, W. A. Knight. R. et al. 2011.
Viladomiu, M., R. Hontecillas, L. Yuan, P. Lu, and J. Bassaganya-Riera. Linking long-Term Dietary patterns with gut microbial enterotypes.
2013. Nutritional protective mechanisms against gut inflammation. Science 334(6052):105–8. doi:10.1126/science.1208344.
International Journal of Biochemistry 24(6):929–39. doi:10.1016/ Wu, Z., R. Guan, M. Tao, F. Lyu, G. Cao, M. Liu, and J. Gao. 2017.
j.jnutbio.2013.01.006. Assessment of the toxicity and inflammatory effects of different-
Vindigni, S. M., T. L. Zisman, D. L. Suskind, and C. J. Damman. 2016. sized zinc oxide nanoparticles in 2D and 3D cell cultures. RSC
The intestinal microbiome, barrier function, and immune system in Advances 7(21):12437–45. doi:10.1039/C6RA27334C.
inflammatory bowel disease: a tripartite pathophysiological circuit Xu, C., C. Yang, Y. Yin, J. Liu, and Y. Mine. 2012. Phosphopeptides
with implications for new therapeutic directions. Therapeutic (PPPs) from hen egg yolk phosvitin exert anti-inflammatory activity
Advances in Gastroenterology 9(4):606–25. doi:10.1177/ via modulation of cytokine expression. Journal of Functional Foods
1756283X16644242. 4(4):718–26. doi:10.1016/j.jff.2012.04.011.
Wallace, D. G., and J. Rosenblatt. 2003. Collagen gel systems for sus- Yasumatsu, H., and S. Tanabe. 2010. The casein peptide Asn-Pro-Trp-
tained delivery and tissue engineering. Advanced Drug Delivery Asp-Gln enforces the intestinal tight junction partly by increasing
Reviews 55(12):1631–49. doi:10.1016/j.addr.2003.08.004. occludin expression in caco-2 cells. British Journal of Nutrition
Walters, W. A., Z. Xu, and R. Knight. 2014. Meta-analyses of human 104(07):951–6. doi:10.1017/S0007114510001698.
gut microbes associated with obesity and IBD. FEBS Letters Yi, J., T. A. Knudsen, A.-L. Nielsen, L. Duelund, M. Christensen, P.
588(22):4223–33. doi:10.1016/j.febslet.2014.09.039. Hervella, D. Needham, and O. G. Mouritsen. 2016. Inhibition of
Wang, F., W. Vallen Graham, Y. Wang, E. D. Witkowski, B. T. cholesterol transport in an intestine cell model by pine-derived phy-
Schwarz, and J. R. Turner. 2005. Interferon-c and tumor necrosis tosterols. Chemistry and Physics of Lipids 200 :62–73. doi:10.1016/
factor-a synergize to induce intestinal epithelial barrier dysfunction j.chemphyslip.2016.06.008.
by up-regulating myosin light chain kinase expression. The Yu, J., S. Peng, D. Luo, and J. C. March. 2012. In vitro 3D human
American Journal of Pathology 166(2):409–19. doi:10.1016/s0002- small intestinal villous model for drug permeability determination.
9440(10)62264-x. Biotechnology and Bioengineering 109(9):2173–8. doi:10.1002/
Wang, F.-C. 2015. Overexpression of HMGB1 A-box reduced lipopoly- bit.24518.
saccharide-induced intestinal inflammation via HMGB1/TLR4 sig- Zachos, N. C., O. Kovbasnjuk, J. Foulke-Abel, J. In, S. E. Blutt, H. R.
naling in vitro. World Journal of Gastroenterology 21(25):7764. De Jonge, M. K. Estes, and M. Donowitz. 2015. Human enteroids/
doi:10.3748/wjg.v21.i25.7764. colonoids and intestinal organoids functionally recapitulate normal
Watson, C. J., C. J. Hoare, D. R. Garrod, G. L. Carlson, and G. intestinal physiology and pathophysiology. The Journal of Biological
Warhurst. 2005. Interferon-c selectively increases epithelial perme- Chemistry 291(8):3759–66. doi:10.1074/jbc.R114.635995.
ability to large molecules by activating different populations of para- Zhang, H., Y. I. Hassan, J. Renaud, R. Liu, C. Yang, Y. Sun, and R.
cellular pores. Journal of Cell Science 118(22):5221–30. doi:10.1242/ Tsao. 2017. Bioaccessibility, bioavailability, and anti-inflammatory
jcs.02630. effects of anthocyanins from purple root vegetables using Mono-
Watt, F. M. 1988. Effect of seeding density on stability of the differenti- and co-culture cell models. Molecular Nutrition & Food Research
ated phenotype of pig articular chondrocytes in culture. Journal of 61(10):n/a–n/a. doi:10.1002/mnfr.20:1600928.
Cell Science 89(3):373–8. Zimmer, J., B. Lange, J.-S. Frick, H. Sauer, K. Zimmermann, A.
WeR glarz, L., J. Wawszczyk, A. Orchel, M. Jaworska-Kik, and Z. Schwiertz, K. Rusch, S. Klosterhalfen, and P. Enck. 2012. A vegan or
Dzier_zewicz. 2007. Phytic acid modulates in vitro IL-8 and IL-6 vegetarian diet substantially alters the human colonic faecal micro-
release from colonic epithelial cells stimulated with LPS and IL-1b. biota. European Journal of Clinical Nutrition 66(1):53–60.
Digestive Diseases and Sciences 52(1):93–102. doi:10.1007/s10620- doi:10.1038/ejcn.2011.141.
006-9320-0. Zoumpopoulou, G., E. Tsakalidou, J. Dewulf, B. Pot, and C. Grangette.
Wehkamp, J., M. Schmid, K. Fellermann, and E. F. Stange. 2005. 2009. Differential crosstalk between epithelial cells, dendritic cells
Defensin deficiency, intestinal microbes, and the clinical phenotypes and bacteria in a co-culture model. International Journal of Food
of Crohn’s disease. Journal of Leukocyte Biology 77(4):460–5. Microbiology 131(1):40–51. doi:10.1016/j.ijfoodmicro.2008.12.037.
doi:10.1189/jlb.0904543. Zucco, F., Batto, A. F. Bises, G. Chambaz, J. Chiusolo, A. Consalvo, R.
Williams, K. M., K. Gokulan, C. E. Cerniglia, and S. Khare. 2016. Size Cross, H. Dal Negro, G. de Angelis, I. Fabre. G. et al. 2005. An
and dose dependent effects of silver nanoparticle exposure on intes- inter-laboratory study to evaluate the effects of medium composition
tinal permeability in an in vitro model of the human gut epithelium. on the differentiation and barrier function of caco-2 cell lines.
Journal of Nanobiotechnology 14(1):62 doi:10.1186/s12951-016-0214- ATLA: Alternatives to Laboratory Animals 33(6):603–18.
9. Zweibaum, A., M. Laburthe, E. Grasset, and D. Louvard. 2011. Use of
Wojtal, K. A., L. Wolfram, I. Frey-Wagner, S. Lang, M. Scharl, S. R. cultured cell lines in studies of intestinal cell differentiation and
Vavricka, and G. Rogler. 2013. The effects of vitamin a on cells of function. In Comprehensive physiology, 223–255. Hoboken: John
innate immunity in vitro. Toxicology in Vitro 27(5):1525–32. Wiley & Sons, Inc.
doi:10.1016/j.tiv.2013.03.013. Zweibaum, A., M. Pinto, G. Chevalier, E. Dussaulx, N. Triadou, B.
Wu, C.-H., H.-W. Huang, J.-A. Lin, S.-M. Huang, and G.-C. Yen. Lacroix, K. Haffen, J.-L. Brun, and M. Rousset. 1985. Enterocytic
2011. The proglycation effect of caffeic acid leads to the elevation of differentiation of a subpopulation of the human Colon tumor cell
oxidative stress and inflammation in monocytes, macrophages and line HT-29 selected for growth in sugar-free medium and its inhib-
vascular endothelial cells. The Journal of Nutritional Biochemistry ition by glucose. Journal of Cellular Physiology 122(1):21–9.
22(6):585–94. doi:10.1016/j.jnutbio.2010.05.002. doi:10.1002/jcp.1041220105.

You might also like