SlideShare a Scribd company logo
1 of 9
1Cell & Tissue Transplantation & Therapy 2014:6
Open Access: Full open access to
this and thousands of other papers at
http://www.la-press.com.
Cell & Tissue
Transplantation & Therapy
Human Prenatal Small Intestine Cells as a Valuable Source of Stem Cells
and Epithelial Cells: Phenotypic and Functional Characterization
Rami Nasrallah, Toai Nguyen, Anasua Kusari, Kelly Burgee and Rafael Gonzalez
DaVinci Biosciences LLC, Costa Mesa, California, USA.
ABSTRACT: As the fastest self-regenerating organ, the intestinal epithelium serves primarily to absorb nutrients and minerals, while its other distinct
functional characteristics include the capacity to proliferate and replicate many lineage precursors. Currently, in vitro studies using small intestine cells are
hindered by the lack of a standard cell culture model.
Here, we characterize primary human prenatal small intestine epithelial cells (HPIEC). The data demonstrate that HPIEC express markers indicative of
stem cells (NOTCH1, SOX9, OLFM4, LGR5), epithelial cells (CK18, CDH1), and immunological and entero-endocrinal cells (DEFA5, GPR-120, GLP-1).
More importantly, HPIEC are shown to secrete GLP-1 and lysozyme, and express sucrase–isomaltase, maltase–glucoamylase, and drug transport function
that is cyclosporin A repressible, which demonstrates functionality. These data indicate that HPIEC are a heterogeneous cell population that contains stem
cells and epithelial cells that may be valuable for various functional, developmental, and pharmacologic studies.
KEYWORDS: in vitro, small intestine cells, characterization, stem cells
CITATION: Nasrallah et al. Human Prenatal Small Intestine Cells as a Valuable Source of Stem Cells and Epithelial Cells: Phenotypic and Functional Characterization.
Cell & Tissue Transplantation & Therapy 2014:6 1–9 doi:10.4137/CTTT.S14354.
RECEIVED: January 17, 2014. RESUBMITTED: March 31, 2014. ACCEPTED FOR PUBLICATION: March 31, 2014.
ACADEMIC EDITOR: David T. Harris, Editor in Chief
TYPE: Original Research
FUNDING: This research was supported by DaVinci Biosciences. The authors alone are responsible for the content and writing of the paper.
COMPETING INTERESTS: Authors disclose no potential conflicts of interest.
COPYRIGHT: © the authors, publisher and licensee Libertas Academica Limited. This is an open-access article distributed under the terms of the Creative Commons
CC-BY-NC 3.0 License.
CORRESPONDENCE: rgonzalez@dvbiosciences.com
Introduction
The small intestine plays a crucial role in digestion and
­nutrient absorption, in constant competition with a plethora
of microbes and their toxins which are part of the normal
intestinal flora. As the physical boundary between the outside
and inside environments, the intestinal epithelium is derived
from stem cells. These stem cells reside at the base of the crypt
and provide all the lineages of epithelial cells, as they con-
tinuously divide, differentiate, and migrate toward the apex of
the villus from where they slough off.1,2
 The epithelium of the
adult mammalian intestine is in constant dynamic interaction
with its underlying mesenchyme controlling direct progenitor
proliferation, lineage commitment, terminal differentiation,
and ultimately cell death.2
Small intestine epithelial cells are potentially a suitable
model for researching inflammatory bowel disease, ­colitis,
intestinal cancer, intestinal infections and more importantly,
toxicity and drug discovery. Although recent research has
described a variety of culture methods,3–7
  no simplified
long-term culture system has been established from human
prenatal intestinal tissue that maintains basic crypt–villus
physiology and metabolic functionality in the differentiated
state. The majority of studies are not performed on primary
intestinal epithelial cells that have been derived directly from
human intestinal tissue. This is in part due to limited access to
viable human intestines and the complexity to isolate, purify,
and propagate primary human intestinal epithelial cells. As
an alternative, commercially available intestinal epithelial cell
lines such as Caco-2 and HT29 are used.8–10
 Caco-2, which
is one of the most widely used and best described intestinal
cell models, is derived from a neoplastic tumor of the large
intestine of a 72-year-old man.11
 Although these cells could
be useful for studies on cell growth, the data obtained from
studies on proliferation and ion transport do not ­necessarily
support the results from the experiments performed with
primary human intestinal epithelial cells.12
 Since the use of
Nasrallah et al
2 Cell & Tissue Transplantation & Therapy 2014:6
these cell lines as tools has limitations,2,5,9,10
 efforts have been
directed at developing alternative sources of human intesti-
nal epithelial cells that maintain their functions and are easy
to work with. Although culture systems of human intestinal
cells have been described,2–9
 an adequate and easily obtain-
able primary cell culture model that is characterized and
demonstrates functionality does not exist. The ideal primary
cell model would contain the complete flora of cells normally
found in the small intestine, be reproducible, expandable, and
functional in vitro.
Here, we present phenotypic, molecular biological, and
functional characterization of primary human prenatal small
intestine epithelial cells (HPIECs) in vitro. These small
intestine cells are a heterogeneous population that is eas-
ily expandable, possesses reproducible phenotypic proper-
ties and may possibly evade the immune response as is the
case in fetal cell microchimerism,13
 which would be suitable
for allogeneic transplant. HPIEC express stem cell markers,
LGR5, SOX9, NOTCH1, and OLFM4;14,15
mature epithe-
lial cell marker, CK18; and innate immune marker, DEFA5.
HPIEC maintain the dynamic balance between proliferation–
differentiation unchanged for several passages. HPIEC also
maintain stem cells and terminally differentiated cells. In
addition, the data demonstrate that HPIEC inducibly secrete
peptide GLP-1  and anti-microbial enzyme lysozyme, and
express P-glycoprotein (Pgp) multi-drug transporter, sucrase–­
isomaltase, and maltase–glucoamylase. Collectively, these
properties suggest that these cells are a potential in vitro
model for drug discovery and development, and possibly in
cell transplantation and tissue engineering studies.
Materials and Methods
Cell culture. HPIEC were isolated from 17- to 18-week-
old pre-natal small intestine tissue made available from elec-
tive medical abortions, with informed consent of donors. The
study and consent procedure were approved by an independent
institutional review board (Ethical & Independent Review
Services IRB 00007807; Study #08103–05). Cell dissocia-
tion solution, culture vessel coating solution, and epithelial
pro-conditioned (D-Pro) medium were all from DV Biolog-
ics, LLC (Costa Mesa, CA). The epithelial pro-conditioned
medium was generated by culturing intestinal mesenchy-
mal cells to 80–90% confluency using DMEM high glucose
(Gibco, Carlsbad, CA) and 10% fetal bovine serum (FBS;
Hyclone, Logan, UT), and then switching the media to low
serum, 2% FBS, to stress the cells. The conditioned media were
harvested after each medium replacement. These ­conditioned
media were then combined 50/50 with basal media ­containing
minimum essential medium (Gibco) supplemented with insu-
lin/transferrin/selenium (BD Biosciences, San Diego, CA),
using 20  ng/ml EGF and 10  ng/ml HGF (Peprotech,
Rocky Hill, NJ), 200  ng/ml choleratoxin and 40  µg/ml
heparin (Sigma-Aldrich, St Louis, MO) as per manufac-
turer’s instruction. All cells used for experiments were tested
for authentication using methods described below. Primary
HPIEC were seeded (1.5–4 × 104
 cells/cm2
) in vessels (25 cm2
,
or 6-well plates) treated with culture vessel coating solution
and cultured in epithelial pro-conditioned medium (DV Bio-
logics). To remove dead cells and cellular debris, the medium
was replaced 24 hours after seeding, so cell clusters could fully
adhere to the growth surface. Afterward, medium change was
performed every two days. To maximize the number of serial
passages, sub-culture was performed when the cells were 80 to
90% confluent (4–8 days).
Growth kinetics. HPIEC were seeded in duplicates at
1.5 × 104
 to 2.0 × 104
 cells/cm2
 in 6-well plates treated with
culture vessel coating solution in epithelial pro-conditioned
medium. The cells were dissociated with cell dissociation solu-
tion every seven days, counted, and sub-cultured at the same
seeding density. Population doubling was calculated using the
formula PD  =  [log10
(N1
)  -  log10
(N0
)]/log10
(2) as previously
described.16
Colony forming unit assay. HPIEC were resuspended
in epithelial pro-conditioned medium and seeded onto
six 10 cm dishes. One dish was seeded with 1.0 × 104
 cells
(control) and the remaining five dishes were seeded with
1.0 × 102
 cells (experimental). The plated cells were incubated
in the CO2
  incubator for two weeks. Culture dishes were
rinsed with PBS, fixed (4% formaldehyde and 1% glutaralde-
hyde in PBS), stained with 1% crystal violet in 10% methanol
(all from Fisher Scientific, Pittsburgh, PA), destained with
water, and air dried. Colonies were quantified using Bio-Rad
Quantity One v. 4.6.6  software and imaging system (Bio-
Rad, Hercules, CA).
Immunocytochemistry. Cells were cultured in 6-well
plates, fixed in phosphate-buffered 4% paraformaldehyde
(PFA) and stored at 4°C. After permeabilization with 0.1% of
Triton X-100 for internal antigens (Promega, Madison, WI)
and blocking with 1% BSA (Sigma-Aldrich) and 10% Nor-
mal Donkey Serum (Jackson Immunoresearch, West Grove,
PA), primary antibodies were applied overnight at 4°C. Cells
were incubated with secondary antibody in blocking buffer for
one hour at room temperature (RT). Cells were counterstained
with DAPI (Molecular Probes, Carlsbad, CA) and mounted
with Fluoromount-G (Southern Biotech, Birmingham, AL).
Primary antibodies used were mouse anti-E-cadherin, rab-
bit anti-Ki67, rabbit anti-ZO1, mouse anti-CK18 (Millipore,
Billerica, MA), and rabbit anti-GPCR GPR-120, mouse anti-
SOX9, and rabbit anti-GPCR GPR49 (anti-LGR5) (Abcam,
Cambridge, England). Secondary antibodies Alexa 488 and
Alexa 594  (Molecular Probes, Carlsbad, CA) were used.
As negative controls corresponding specific non-immune
immunoglobulins (Santa Cruz Biotech, Santa Cruz, CA)
were used for all experiments. Staining was analyzed using an
Olympus IX81 inverted microscope and SlideBook 4.2 soft-
ware (Intelligent Imaging Innovations, Inc.).
PCR analysis. RNA was directly isolated from primary
or cultured HPIEC and brain tissue using the RNeasy Mini
Human prenatal small intestine cells as a valuable source stem and epithelial cells
3Cell & Tissue Transplantation & Therapy 2014:6
Kit (Qiagen, Hilden, Germany). cDNA was synthesized
with Superscript III (Invitrogen, Carlsbad, CA) using ran-
dom hexamers and 200 ng of total RNA in a 25-µl reaction.
PCR was conducted in a C1000TM
Thermal Cycler (Bio-Rad,
Hercules, CA) using GoTaq® Green Master Mix (Promega)
and 2 µl of cDNA product for the analyses of all genes. No RT
controls were included in each experiment. RNA from brain
tissue was used as a control to show expression of these genes
in another tissue type. PCR products were visualized by aga-
rose gel electrophoresis. Table 1 lists PCR primer sequences
used for all experiments.
Lysozyme functional assay. The lysozyme assay was
performed according to manufacturer’s protocol (Worthing-
ton Biochemical, Lakewood, NJ) with modifications. Briefly,
HPIEC were incubated in triplicates for two hours in assay
buffer alone DMEM high glucose (Invitrogen) plus 15% FBS
(HyClone) and as applicable supplemented with 0.3  mg/
ml Micrococcus lysodeikticus. The clarified supernatants were
assayed for their ability to lyse a suspension of M. lysodeikticus
over time, by recording the absorbance at 450 nm every four
­minute; a solution of 5 U/ml purified lysozyme (Worthing-
ton Biochemical) in assay buffer was processed in parallel as
a control.
GLP-1  assay. ELISA was performed according to the
manufacturer’s suggested protocol for GLP-1 assay kit (Mil-
lipore). Briefly, HPIEC were cultured in duplicates in 12-well
plates for four days. The cells were washed twice with a rinse
buffer (PBS supplemented with CaCl2
  and MgCl2
, 2  and
1 mmol/l,respectively,and 0.5% BSA),incubated for two hours
at 37°C, 5% CO2
, in 500 µl rinse buffer containing 0.1 mmol/l
Diprotin A (Sigma-Aldrich), supplemented with glucose (5 or
25 mmol/l); or with tolbutamide and/or forskolin and isobutyl-
methylxanthine (IBMX) (10, 10, and 500 µmol/l, respectively).
Tolbutamide, forskolin, and IBMX (all from Sigma-Aldrich)
were diluted from 1000× stock solutions in DMSO. Solu-
tions with equivalent final DMSO concentrations were used as
controls for samples containing tolbutamide, and/or forskolin
and IBMX. GLP-1 in clarified supernatants was quantified by
ELISA using a VMax ­Microplate Reader (Molecular Devices,
Sunnyvale, CA).
Multidrug transporter assay. The experiment was per-
formedusingVybrant®MultidrugResistanceAssayKitaccord-
ing to the manufacturer’s suggested protocol (Invitrogen).
Briefly, HPIEC and Caco-2 cells were incubated in growth
medium (DMEM supplemented with 15% FBS) with 0, 0.165,
1.48, 13.3, and 120 mg/l cyclosporin A for 15 min, exposed
Table 1. Primer sequences used in PCR studies.
GENE SEQUENCE AMPLICON SIZE
CK14
ACGATGGCAAGGTGGTGT
118 bp
GGGATCTTCCAGTGGGATCT
DEFA5
TGAGGACCATCGCCATCCTTGCT
226 bp
TCACGGGTAGCACAACGGCCG
GAPDH
CAAGGTCATCCATGACAACTTTG
496 bp
GTCCACCACCCTGTTGCTGTAG
OLFM4
CCAGCTAAGAGGACAAGATGAG
286 bp
GGCAGGGAAACAGAGCAC
LGR5
GAACATGCTCACGGGAGTCTC
303 bp
CCACCCAGCAGGGGAAC
CK18
GGGAGCACTTGGAGAAGAAG
229 bp
GATATTGGTGTCATCAATGACCT
E-cadherin-1
AGGAACACAGGAGTCATCAGTG
260 bp
GGGGTATTGGGGGCATC
GRP120
GCTCTGCCTCTCTGCGTCT
255 bp
GCGGATCTGGTGGCTCTC
GLP1
ATCATTCTCAGCTTCCCAGG
370 bp
CTCATCAGAGAAAGAACCATCAG
NOTCH-1
CGCCTTTGTGCTTCTGTTC
300 bp
GGTGGTCTGTCTGGTCGTC
Sucrase Isomaltase
TTCGCTACACCTTATTACCCTTCC
415 bp
TCTCCTTTGGCTGTGTTGTTTTC
Maltase Glucoamylase
CCCTTCGCCTGGATGTCAC
371 bp
TGGGGGCTGGTCTCGG
Nasrallah et al
4 Cell & Tissue Transplantation & Therapy 2014:6
to 0.25 µmol/l calcein acetoxymethyl ester (calcein-AM) for
15 min, washed twice with growth medium, and the fluores-
cence was quantified using the Bio-Rad CFX-96 RT system.
The results were normalized to no cyclosporin A control.
Results
Propagation of HPIEC. Since there is a current absence
of a small intestine primary cell model, we took on the task
to characterize small intestine cells isolated from prenatal
­tissue and propagate them using a conditioned media obtained
from intestine-derived mesenchymal cells. The culture sys-
tem described herein has been optimized specifically for the
culture of small intestinal epithelial cells as multi-cellular
aggregates (organoids), since previous experimentation with
intestinal epithelial cells indicated that breaking down the
epithelium to a single-cell suspension led to poor adherence
and survival of the epithelial cells. Initially seeded as organ-
oids (Fig. 1A), HPIEC exhibit better long-term viability in
culture in comparison to starting from a single-cell suspension
(personal observation, data not shown). In D-PRO medium,
these organoids quickly formed a lawn of highly enriched
small intestine epithelial cells (Fig. 1B). Easily sub-cultured
with the use of cell dissociation solution, HPIEC maintained
their phenotypical morphology after four passages or nine
population doublings (Fig. 1C). HPIEC could be propagated
for at least six passages or 14 population doublings without
any noticeable morphological changes (Fig. 1D).
HPIEC characterization. To better describe HPIEC,
we performed molecular and immunological characteriza-
tions using immunocytochemistry (ICC) and RT-PCR.
ICC analysis demonstrates that HPIEC express division and
­proliferative marker Ki67 (Fig. 2A). Similar to Ki67 ICC stain-
ing, the stem cell marker SOX9 is also expressed in some of
the HPIEC (Fig. 2A). This may explain why HPIEC are eas-
ily expandable. The staining of both Ki67 and SOX9 appears
to be confined to the inner cells of epithelial colonies that
are formed.
Interestingly, within organoids, the epithelial stem cell
marker LGR-5 is also found (Fig. 2B). Our RT-PCR data
support and confirm our ICC results. PCR analysis demon-
strates that HPIEC express early development and stem cell
markers NOTCH1, SOX9, OLFM4, and LGR5 (Fig. 2C).
0 10 20 30 40
0.0
3.0
6.0
9.0
12.0
15.0
Populationdoubling
Days in culture
D
Figure 1. Growth characteristics of HPIEC. A) Phase contrast of freshly isolated HPIEC. After overnight attachment HPIEC show characteristic
organoid structure of intestinal epithelial cells. B) Established culture of HPIEC, Passage 0 phase contrast. After 7 days in culture in epithelial pro
conditioned medium, HPIEC display typical morphology of intestinal epithelial cells. C) Established culture of HPIEC, Passage 4 phase contrast. After
over 8 population doublings in culture using epithelial pro conditioned medium, HPIEC maintain typical morphology of intestinal epithelial cells. D) HPIEC
growth curve illustrating cumulative population doublings.
Human prenatal small intestine cells as a valuable source stem and epithelial cells
5Cell & Tissue Transplantation & Therapy 2014:6
In addition, we performed colony forming unit (CFU)
assays to confirm the presence of stem cells. When HPIEC
were seeded at 100  cells per 10  cm dish, the data illus-
trate the formation of 9.6  ±  0.7  CFUs (n  =  5). Control
that was seeded at 10,000 cells per 10 cm dish produced
a lawn of cells at two weeks (data not shown). HPIEC
express markers indicative of mature epithelial cells.
HPIEC express adhesion of epithelial cell marker E-cad-
herin, tight junction associated protein marker ZO-1, the
mature epithelial marker CK18, and intestinal marker G
protein coupled receptor 120 (GPR-120) (Fig. 3A). Our
PCR data (Fig. 3B) support and extend the ICC results.
HPIEC express mature epithelial cell markers CK18 and
E-cadherin, and also express the Paneth cell marker
defensin-α-5 (DEFA5), which is thought to be involved in
host defense. In addition, the markers GPR-120 and glu-
cagon-like peptide 1 (GLP-1), which are thought to play
an important role in regulating insulin, are also expressed
(Fig. 3B). Indeed, our PCR analysis of epithelial cell
markers (CK-18 and CDH1), epithelial stem cells markers
(LGR5, SOX9, and OLFM4), and Paneth cells (DEFA5)
demonstrates that cultured cells in conditioned medium
maintain the same gene expression profile as uncultured
freshly isolated cells (Figs. 2 and 3, lane 2). These data sup-
port the notion that HPIEC contain a mixed population of
stem cells and terminally differentiated cells.
HPIECfunctionality.We observed that HPIEC secrete
GLP-1  and lysozyme upon stimulation and express a
repressible multidrug transporter function (Fig. 4). Using
­enzyme-linked immunosorbent assay (ELISA), we found
that HPIEC secrete GLP-1, a protein synthesized only in
the intestine by posttranslational processing of proglucagon
via the L-cell.17
 HPIEC secrete GLP-1 in response to glucose
in a dose-dependent manner and upon stimulation with the
anti-diabetes compound tolbutamide and elevated cAMP lev-
els that are induced by forskolin and IBMX (Fig. 4A).
Paneth cells secrete lysozyme and α-defensins, both of
which have clear antimicrobial activity, and also tumor necro-
sis factor-alpha, and phospholipase A2.18 
For further func-
tional testing,HPIEC were exposed to M. Lysodeikticus,which
stimulated the secretion of lysozyme (Fig. 4B), thus confirm-
ing that the present culture system preserves the subpopula-
tion diversity comprising Paneth cells, among others that
exists in the natural environment of the small intestine. We
also researched for the presence of Pgp multi-drug transporter
activity in HPIEC. This was determined using the Invitrogen
Vybrant MDR assay kit. This assay kit relies on the passive
diffusion of the lipophilic calcein-AM, which can be carried
outside the cell by a Pgp multi-drug transporter, if it is not
converted to fluorescent calcein by indigenous esterases. The
Pgp multi-drug transporter can be inhibited by cyclosporin
A, resulting in an intracellular accumulation of fluorescent
calcein. The data demonstrate that, similar to Caco-2 cells, in
the present culture system, HPIEC express a Pgp ­multi-drug
transporter activity, which is inhibited by cyclosporin A in a
dose-dependent manner (Fig.4C).Our PCR data ­demonstrate
A
B C 1 2 3 4
NOTCH1
SOX9
OLFM4
LGR5
GAPDH
Figure 2. Immunocytochemical staining and Molecular marker characteristics of HPIEC. HPIEC were stained with specific monoclonal or
polyclonal antibodies, counter-stained with DAPI (Blue), and visualized with fluorescence microscopy. Panel A illustrates staining of HPIEC with
antibodies specific for SOX9 (green) and Ki67 (red). Merged images demonstrate that not all cells express SOX9 and Ki67. Panel B illustrates staining
of LGR5 which is mostly confined to organoid structures. For panel C total RNA was isolated and reverse transcribed (RT); the resulting cDNA was PCR-
amplified with primer pairs specific for markers of stem cells NOTCH1, SOX9, OLFM4, and LGR5. GAPDH was included as housekeeping marker control.
Lane 1: No RT control; lane 2: Uncultured HPIEC; lane 3: Passage 4 HPIECs; lane 4: Brain tissue.
Nasrallah et al
6 Cell & Tissue Transplantation & Therapy 2014:6
that HPIEC express sucrase–isomaltase and ­maltase–glu-
coamylase (Fig. 4D).The expression of specific enzyme mark-
ers sucrase–isomaltase and maltase–­glucoamylase is ­indicative
of the presence of functional entrocytes. ­Moreover, our PCR
analysis demonstrates that cultured cells in conditioned
medium maintain the same expression profile as uncultured
freshly isolated cells (Figs. 4D, lane 2  and  3). These results
further support the notion that HPIEC are functional and a
valuable source for various studies.
Discussion
Recent advances in small intestine research illustrate the
intimate interaction between intestinal stem cells and the
self-renewing epithelium. This interaction can be explained
by an intricate interplay among a distinct set of growth sig-
nals that are now becoming well defined.15,19
 Presently, there
is no standard in vitro cell culture system for small intes-
tine epithelial cells although several have been reported in
the literature.5–7,19
 Sato et al described a small intestine cell
culture system that is in a 3D matrix and requires organoid
formation and maintenance. Although this system may have
limitless potential, it does not appear to be a simplified in
vitro model and to the best of our knowledge they have yet
to test the ability of this model for specific in vitro studies
such as GLP-1  secretion and multi-drug transporter assays
in human cells.19
The purpose of this study was to identify a
simple cell culture system of small intestine epithelial cells
that mimics the in vivo system. The system must be easy to
work with and should be valuable for multiple in vitro studies.
After researching current literature, we decided to use cells
isolated from prenatal small intestine. HPIEC are a hetero-
geneous monolayer cell population that contains all the cell
populations of the small intestine. Monolayer of cells is widely
used throughout industry and enables rapid research and drug
discovery in vitro. HPIEC exhibit stem cell self-renewal and
multipotential differentiation in culture. In addition, the close
proximity of Paneth cells to stem cell populations provides
intestinal epithelial cells with essential signals to assemble
into self-organizing structures. Thus, it is imperative to take
into consideration isolation techniques that preserve close cel-
lular interactions.3
The culture vessel coating solution and conditioned
media of the presently described culture system provides an
adequate environment supporting the growth of HPIEC.
Current evidence illustrates the significant roles of the extra-
cellular matrix (ECM) as well as cell culture media on the
attachment, differentiation, and proliferation of intestinal
epithelia.3,4,20–22
 Since the proliferation of epithelial crypt is
Wnt dependent,23,24
 the method employed herein also simu-
lates the extracellular signals found in the intestinal crypt, by
utilizing a medium conditioned by mesenchymal and Paneth
cells that reside in the crypt–villus niche. These cells secrete
various cytokines that modulate the Wnt gene expression
pathway similar to that expressed in vivo.23,24
Although the growth, maintenance, and propagation of
small intestinal epithelial cells can be challenging, HPIEC are
easily expandable and maintain their phenotype (Fig. 1), most
likely due to the method of isolating ­organoids consisting of
intact villi and crypt units that have a much greater capacity to
adhere, survive, and maintain the polarity of epithelium.3
 Base
columnar cells in select populations of SOX9 expressing cells
in crypt cells were shown to produce multilineage ­intestinal
organoids in ex vivo culture systems.25
  Indeed, HPIEC
express dividing cell marker Ki67  and stem cell markers
A
B 1 2 3 4
CK18
CDH1 (E-cadherin)
DEFA5
GPR-120
GLP-1
GAPDH
Figure 3. Immunocytochemical staining and Molecular marker
characteristics of HPIEC. HPIEC were stained with specific monoclonal
or polyclonal antibodies, counter-stained with DAPI (Blue), and visualized
with fluorescence microscopy. Panel A illustrates antibodies specific for
E-cadherin (green), ZO-1 (red), cytokeratin-18 (CK18; green) and
G protein-coupled receptor 120 (GPR-120; red). For panel B total RNA
was isolated and reverse transcribed (RT); the resulting cDNA was
PCR-amplified with primer pairs specific for epithelial cells (CK18,
CDH1), and entero-immunological and entero-endocrinal characteristics
(DEFA5, GPR-120, GLP-1); GAPDH was included as housekeeping
marker control. Lane 1: No RT control; lane 2: Uncultured HPIEC;
lane 3: Passage 4 HPIECs; lane 4: Brain tissue.
Human prenatal small intestine cells as a valuable source stem and epithelial cells
7Cell & Tissue Transplantation & Therapy 2014:6
*
*
*
Control
Tolbutamide
[GLP-1]inmedium(pM)
Forskolin+IBMX
Tolb+Forsk+IBMX
Glucose5mM
Glucose25mM
0
20
40
60
A
A450
Elapsed time (min)
0.00
0.20
0.40
0.60
0.80
1.00
0 5 10 15 20 25
B
*
**
*
*
*
0.000
0.165
1.481
13.333
80
120
160
200
240
%retention
µg/ml Cyclosporin A
C
D 1 2 3 4
Sucrase-isomaltase
Maltase-glucoamylase
GAPDH
Figure 4. Functional characterization of HPIEC. A) Synthesis of glucagon-like peptide 1 (GLP-1). PD015-F cells were equilibrated in assay buffer
for 2 h, then exposed to tolbutamide, or forskolin and isobutylmethylxanthine (IBMX), or tolbutamide, forskolin and IBMX, or 5 mM glucose, or 25 mM
glucose as indicated for 2 h. GLP-1 in the clarified supernatants was assayed by ELISA, and normalized to no treatment control. B) Synthesis of
lysozyme. PD015-F cells were incubated in assay buffer alone (¡), or challenged with 0.3 mg/ml Micrococcus lysodeikticus (ML; ¨) for 2 h. The ability of
the clarified supernatants to lyse a suspension of ML over time was assayed; a solution of 5 U/ml purified lysozyme served as positive control (∆).
C) Inhibition of P-glycoprotein activity. PD015-F cells (Lighter shade) and Caco-2 cells (Darker shade) were treated in various concentrations of
cyclosporin A for 15 min, and loaded with calcein-AM (0.25 µmol/l) for 15 min; after two washes, and residual fluorescence was quantified, and normalized
to no cyclosporin A control. D) Expression of sucrase-isomaltase and maltase-glucoamylase. Total RNA was isolated and reverse transcribed (RT);
the resulting cDNA was PCR-amplified with primer pairs specific for sucrase-isomaltase and maltase-glucoamylase. Lane 1: No RT control; lane 2:
Uncultured HPIEC; lane 3: Passage 4 HPIECs; lane 4: Brain tissue. Error bar: ± SEM. Asterix (*) denotes statistically significant difference over control.
SOX9, NOTCH1, OLFM4 and LGR5 (Fig. 2). Colony form-
ing unit assay of HPIEC suggest that approximately 9.6% of
the cells may be of the stem cell origin (data not shown). The
co-localization of SOX9 and the proliferation marker Ki67 in
cultured cells (Fig. 2) illustrate the possibility of using human
fetal cells to isolate selective stem populations capable of gen-
erating complete villi and crypt in vitro for tissue engineering
purposes. Figure 2 also demonstrates the expression of LGR5,
a Wnt targeting gene that is selectively expressed in the base
of the intestinal crypt. LGR5+ stem cells are critical regula-
tors of the cell populations at the crypt base. LGR5+ cells are
unique due to their capability to form self-renewing, and mul-
tipotent organoids that can recapitulate the structure and cel-
lular organization of intestinal epithelium from a single cell.19
HPIEC are a monolayer of cells. They consist of a het-
erogeneous population of cells found in the small intestine
(Figs. 1–3). HPIEC express markers indicative of mature epi-
thelial cells (E-cadherin, ZO-1, CK18; Fig. 3), and immuno-
logical and entero-endocrinal cell markers (DEFA5, GPR-120,
GLP-1; Fig. 3). These data provide evidence of a natural diver-
sity of native subpopulations of ­progenitor cells, stem cells,
and their progeny of transit-amplifying cells.2,3,16
  HPIEC
take advantage of normal stem cell properties related in
organoids.19
 Future studies will aim to identify percentages of
specific cell types within the HPIEC population.
Several commercially available intestinal epithelial cell
lines are being used for functional studies. These commer-
cially available cell lines are derived from transformed human
Nasrallah et al
8 Cell & Tissue Transplantation & Therapy 2014:6
­tissue such as polyps and cancerous lesions (HT-29, Caco-
2, and T84),8–10
  which have their limitations that include
their malignant nature and colonic origin.5,8
  HPIEC are
a monolayer cell system that may be similar to native small
intestine, which is confirmed by the expression of specific
intestinal metabolite enzymes and transporter proteins such
as ATP-binding cassette (ABC) transporters Pgp expressed
by absorptive enterocytes. Since HPIEC maintain mature and
specialized intestinal cell populations after isolation and prop-
agation, these cells provide researchers with a tool to inves-
tigate digestive and absorptive capacities of normal human
intestines (Fig. 4).
HPIEC have the ability to secrete GLP-1 upon stimula-
tion with glucose or anti-diabetic and cAMP-elevating com-
pounds (Fig. 4). GLP-1 is a secretory peptide of physiological
importance. It acts as a potent incretin, specifically stimulat-
ing glucose-induced insulin release, inhibiting glucagon secre-
tion, and modulating the ileal control machinery by slowing
down gastric emptying.17,26
 As a major regulator of the insu-
lin secretory pathway, GLP-1  has many vital physiologi-
cal actions, with many great implications in recent diabetes
research.16
 In addition, it has been shown to affect the central
nervous system, with its neuroprotective properties and influ-
ence on learning and memory.26
 Another important feature of
GLP-1 is that it plays an inhibitory role in the regulation of
eating and drinking.27
 GLP-1 or a potent, long-acting natu-
ral or synthetic GLP-1  agonist, such as Exenatide (Byetta)
or Liraglutide, were demonstrated to lower glycemia which
has resulted in its acceptance as a treatment for patients with
type 2 diabetes.28
 These data suggest that HPIEC may be a
potential research model in the areas of gastrointestinal and
neurological disorders, and diabetes therapy.
HPIECsecretelysozymeuponexposuretoM.lysodeikticus
(Fig. 4) which illustrates that these cells preserve Paneth cell
functional properties in the context of the intestinal epithe-
lium. Paneth cells, which are characterized by their large
eosinophilic granules of anti-microbial compounds, comprise
another important subpopulation of the intestinal epithelium.
They are located near the crypts that secrete, among other
substances, defensin-α and lysozyme, following interaction
with bacterial antigens and in response to pharmacologic
stimulation.18,29,30
 It has been shown that there is decreased
expression of α-defensins in Crohn’s iletis.31
 Since HPIEC
contain Paneth cells, this may serve as a model for research in
intestinal immunity, encompassing inflammatory bowel dis-
ease, Crohn’s disease, and other ­gastrointestinal disorders.31
Shown to express a Pgp-like function, repressible
by cyclosporin A, using calcein-AM as the test substrate,
HPIEC may be a suitable model for rapid screening of intesti-
nal drug absorption and the detection of chemical compounds
­interfering with Pgp-like functions (Fig. 4C). Intestinal epi-
thelium has the ability to transport toxic substances, such as
chemotherapy drugs, which may have been taken up non-
specifically by passive diffusion or in any other way into the
lumen. In some cancers, these efflux functions are highly
amplified and contribute to their resistance to chemotherapy.
The intestinal epithelial native non-specific efflux function is a
current subject in the screening process for suitable anti-cancer
drugs that can circumvent the potential problem of resistance.
Pgp, the most widely studied ABC transporter, functions
as a biological barrier by regulating the cellular accumula-
tion and disposition of cytotoxic and xenobiotics drugs out of
cells.32,33
 In vitro and in vivo studies have demonstrated that
Pgp is involved in the export of a variety of natural drugs and
lies within the luminal membranes of the endothelial cells
of the brain and testes, in the adrenal glands, renal proximal
tubule cells, and in the ­apical membrane of epithelial cells of
the liver, colon, and small intestine.
Responses to external stimuli have been poorly studied in
most in vitro culture methods; however, the data demonstrated
here show that HPIEC have the indicative feature to respond
to stimuli in vitro. The expression of specific enzyme markers
sucrase–isomaltase and maltase–glucoamylase is indicative of
the presence of functional enterocytes (Fig. 4D). Enterocytes
are responsible for many cytokines secretion, inflammatory
response, and play a role in microflora adhesion proteins as
well as bacterial pathogenesis.11
 Enterocytes play a key role
in responses to external stimuli and interaction with the gut’s
bacterial flora. Thus, cytokine expressing cells are important
when considering assembly of responsive architectural model
of intestinal tissue.
In summary, here we have characterized a prenatal, het-
erogeneous primary small intestine epithelial cell which we
designate HPIEC, which express several stem cell and mature
epithelial cell markers. More importantly, the data illus-
trate that HPIEC may be an appropriate and adequate cell
source for in vitro research in gastrointestinal disorders, dia-
betes research, drug discovery and tissue engineering studies
since they contain stem cells (LGR5+), enterocytes (sucrase–­
isomaltase and maltase–glucoamylase expression), Paneth cells
(lysozyme secretion), and enteroendocrine cells (GLP-1 secre-
tion). Future studies will aim to quantify specific cell types
in HPIEC and target growing the cells in a 3D environment
or using biodegradable matrices in order to reconstitute the
small intestine environment. Such studies precipitate research
to help individuals suffering from intestinal cancers and pos-
sibly deficiencies of sucrase–isomaltase.
Acknowledgements
Authors are thankful to Dr. Nickolas Chelyapov and
Dr. Robert Sackstein for manuscript review and suggestions.
Author Contributions
Conceived and designed the experiments: RN, TN, RG.
Analyzed the data: RN, TN, RG. Wrote the first draft of
the manuscript: RN, RG. Contributed to the writing of the
­manuscript: TN, AK, KB. Agree with manuscript results
and conclusions: RN, TN, AK, KB, RG. Jointly developed
Human prenatal small intestine cells as a valuable source stem and epithelial cells
9Cell & Tissue Transplantation & Therapy 2014:6
the structure and arguments for the paper: RN, TN, RG. All
authors reviewed and approved of the final manuscript.
DISCLOSURES AND ETHICS
This paper was subject to independent, expert peer review by a minimum of two blind
peer reviewers. All editorial decisions were made by the independent academic edi-
tor. All authors have provided signed confirmation of their compliance with ethical and
legal obligations including (but not limited to) use of any copyrighted material, com-
pliance with ICMJE authorship and competing interests disclosure guidelines and,
where applicable, compliance with legal and ethical guidelines on human and animal
research participants.
REFERENCES
	 1.	 Dignass AU, Tsunekawa S, Podolsky DK. Fibroblast growth factors modulate
intestinal epithelial cell growth and migration. Gastroenterology. 1994;106:
1254–1262.
	 2.	 Chopra DP, Dombkowski AA, Stemmer PM, Parker GC. Intestinal epithelial
cells in vitro. Stem Cells Dev. 2010;19:131–142.
	 3.	 Evans GS, Flint N, Somers AS, Eyden B, Potten CS. The development of a
method for the preparation of rat intestinal epithelial cell primary cultures. J Cell
Sci. 1992;101:219–231.
	 4.	 Perreault N, Beaulieu J. Primary cultures of fully differentiated and pure human
intestinal epithelial cells. Exp Cell Res. 1998;245:34–42.
	 5.	 Pageot LP, Perreault N, Basora N, Francoeur C, Magny P, Beaulieu JF. Human
cell models to study small intestinal functions: recapitulation of the crypt-villus
axis. Microsc Res Tech. 2000;49:394–406.
	 6.	 Liu Z, Zhang P, Zhou Y, Qin H, Shen T. Culture of human intestinal epithelial
cell using the dissociating enzyme thermolysin and endothelin-3. Braz J Med Biol
Res. 2010;43:451–459.
	 7.	 Sato T. Novel intestinal stem cell culture system. Inflamm Regen. 2012;32:43–47.
	 8.	 Rousset M. The human colon carcinoma cell lines HT-29  and Caco-2: two
in vitro models for the study of intestinal differentiation. Biochimie. 1986;68:
1035–1040.
	 9.	 Parlesak A, Haller D, Brinz S, Baeuerlein A, Bode C. Modulation of cytokine
release by differentiated CACO-2 cells in a compartmentalized coculture model
with mononuclear leucocytes and nonpathogenic bacteria. Scand J Immunol.
2004;60:477–485.
	10.	 Huang DB, DuPont HL, Jiang ZD, Carlin L, Okhuysen PC. Interleukin-8
response in an intestinal HCT-8 cell line infected with enteroaggregative and
enterotoxigenic Escherichia coli. Clin Diagn Lab Immunol. 2004;11:548–551.
	 11.	 Grajek W, Olejnik A. Epithelial cell cultures in vitro as a model to study func-
tional properties of food. Pol J Food Nutr Sci. 2004;1(3/54):5–24.
	 12.	 Jung HC, Eckmann L, Yang SK, et al. A distinct array of proinflammatory cyto-
kines is expressed in human colon epithelial cells in response to bacterial inva-
sion. J Clin Invest. 1995;95:55–65.
	13.	 Bianchi DW, Zickwold GK, Weil GJ, Sylvester S, DeMaria MA. Male fetal
progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc
Natl Acad Sci U S A. 1996;93(2):705–708.
	14.	 Formeister EJ, Sionas AL, Lorance DK, Barkley CL, Lee GH, Magness ST.
Distinct SOX9  levels differentially mark stem/progenitor populations and
enteroendocrine cells of the small intestine epithelium. Am J Physiol Gastrointest
Liver Physiol. 2009;296:G1108–G1118.
	15.	 Sato T, van Es JH, Snippert HJ, et al. Paneth cells constitute the niche for
LGR5 stem cells in intestinal crypts. Nature. 2011;469:415–418.
	 16.	 Cristofalo VJ, Allen RG, Pignolo RJ, Martin BG, Beck JC. Relationship between
donor age and the replicative lifespan of human cells in cultura: a reevaluation.
Proc Natl Acad Sci U S A. 1998;95:10614–10619.
	 17.	 Holst JJ. Enteroglucagon. Annu Rev Physiol. 1997;59:257–271.
	 18.	 Peeters T, Vantrappen G. The Paneth cell: a source of intestinal lysozyme. Gut.
1975;16:553–558.
	 19.	 Sato T, Vries RG, Snippert HJ, et al. Single LGR5 stem cells build crypt-villus
structures in vitro without a mesenchymal niche. Nature. 2009;459:262–266.
	 20.	 Goke M, Zuk A, Podolsky DK. Regulation and function of extracellular matrix
intestinal epithelial restitution in vitro. Am J Physiol Gastrointest Liver Physiol.
1996;271:G729-G740.
	 21.	 Sanderson IR, Ezzell RM, Kedinger M, et al. Human fetal enterocytes in vitro:
modulation of the phenotype by extracellular matrix. Proc Natl Acad Sci U S A.
1996;93:7717–7722.
	 22.	 Hofmann C, Obermeier F, Artinger M, et al. Cell–cell contacts prevent anoikis
in primary human colonic epithelial cells. Gastroenterology. 2007;132:587–600.
	 23.	 Kuhnert F, Davis CR, Wang HT, et al. Essential requirement for Wnt signaling
in proliferation of adult small intestine and colon revealed by adenoviral expres-
sion of Dickkopf-1. Proc Natl Acad Sci U S A. 2004;101:266–271.
	 24.	 Ootani A, Li X, Sangiorgi E, et al. Sustained in vitro intestinal epithelial culture
within a Wnt-dependent stem cell niche. Nat Med. 2009;15:701–706.
	25.	 Barker N. Adult intestinal stem cells: critical drivers of epithelial homeostasis
and regeneration. Nat Rev Mol Cell Biol. 2014;15:19–33.
	26.	 Harkavyi A, Whitton PS. Glucagon-like peptide 1  receptor stimulation as a
means of neuroprotection. Br J Pharmacol. 2010;159:495–501.
	 27.	 Hayes MR, De Jonghe BC, Kanoski SE. Role of the glucagon-like-peptide-1
receptor in the control of energy balance. Physiol Behav. 2010;100:503–510.
	 28.	 Bond A. Exenatide (Byetta) as a novel treatment option for type 2 diabetes mel-
litus. Proc (Bayl Univ Med Cent). 2006;19(3):281–284.
	 29.	 Wilson CL, Ouellette AJ, Satchell DP, et al. Regulation of intestinal α-defensin
activation by the metalloproteinase matrilysin in innate host defense. Science.
1999;286:113–117.
	 30.	 Tanabe H, Ayabe T, Bainbridge B, et al. Mouse Paneth cell secretory responses
to cell surface glycolipids of virulent and attenuated pathogenic bacteria. Infect
Immun. 2005;73:2312–2320.
	31.	 Elphick DA, Mahida YR. Paneth cells: their role in innate immunity and
inflammatory disease. Gut. 2005;54(12):1802–1809.
	 32.	 Gottesman MM, Pastan I. Biochemistry of multidrug resistance mediated by the
multidrug transporter. Annu Rev Biochem. 1993;62:385–427.
	 33.	 Cole SP, Sparks KE, Fraser K, et al. Pharmacological characterization of multi-drug
resistant MRP-transfected human tumor cells. Cancer Res. 1994;54: 5902–5910.

More Related Content

What's hot

Nurational requirment growth and metabolism of cell
Nurational requirment growth and metabolism of cellNurational requirment growth and metabolism of cell
Nurational requirment growth and metabolism of cella fatima
 
Liver tissue engineering
Liver tissue engineeringLiver tissue engineering
Liver tissue engineeringKollaSrivalli
 
Primary and established cell line culture
Primary and established cell line culturePrimary and established cell line culture
Primary and established cell line cultureKAUSHAL SAHU
 
Animal cell culture introduction and equipments
Animal cell culture introduction and equipmentsAnimal cell culture introduction and equipments
Animal cell culture introduction and equipmentsDrsajadHassanWani
 
Basic of animal cell culture part iii
Basic of animal cell culture part iiiBasic of animal cell culture part iii
Basic of animal cell culture part iiiFahad Khan
 
Primary culture and cell line
Primary culture and cell linePrimary culture and cell line
Primary culture and cell lineKAUSHAL SAHU
 
Notes for Cell Culture Basic Techniques
Notes for Cell Culture Basic TechniquesNotes for Cell Culture Basic Techniques
Notes for Cell Culture Basic Techniquesimprovemed
 
Cell lines and maintanence
Cell lines and maintanenceCell lines and maintanence
Cell lines and maintanencepaaruselvam
 
Animal cell culture techniques
Animal cell culture techniques Animal cell culture techniques
Animal cell culture techniques Nagendra P
 
Substrates for animal cellculture
Substrates for animal cellcultureSubstrates for animal cellculture
Substrates for animal cellcultureVipin Shukla
 
Cell lines seminar
Cell lines seminarCell lines seminar
Cell lines seminarvarshidora
 

What's hot (20)

Nurational requirment growth and metabolism of cell
Nurational requirment growth and metabolism of cellNurational requirment growth and metabolism of cell
Nurational requirment growth and metabolism of cell
 
Liver tissue engineering
Liver tissue engineeringLiver tissue engineering
Liver tissue engineering
 
3
33
3
 
Stem cells , culture and technologies
Stem cells , culture and  technologiesStem cells , culture and  technologies
Stem cells , culture and technologies
 
Arsh copy
Arsh   copyArsh   copy
Arsh copy
 
Mortal and immortal cell lines
Mortal and immortal cell linesMortal and immortal cell lines
Mortal and immortal cell lines
 
Primary and established cell line culture
Primary and established cell line culturePrimary and established cell line culture
Primary and established cell line culture
 
Cell lines
Cell linesCell lines
Cell lines
 
Animal cell culture introduction and equipments
Animal cell culture introduction and equipmentsAnimal cell culture introduction and equipments
Animal cell culture introduction and equipments
 
Basic of animal cell culture part iii
Basic of animal cell culture part iiiBasic of animal cell culture part iii
Basic of animal cell culture part iii
 
Animal cell introduction
Animal cell introductionAnimal cell introduction
Animal cell introduction
 
Cell lines
Cell lines Cell lines
Cell lines
 
Primary culture and cell line
Primary culture and cell linePrimary culture and cell line
Primary culture and cell line
 
Notes for Cell Culture Basic Techniques
Notes for Cell Culture Basic TechniquesNotes for Cell Culture Basic Techniques
Notes for Cell Culture Basic Techniques
 
Animal cell culture
Animal cell cultureAnimal cell culture
Animal cell culture
 
Cell lines and maintanence
Cell lines and maintanenceCell lines and maintanence
Cell lines and maintanence
 
Animal cell culture techniques
Animal cell culture techniques Animal cell culture techniques
Animal cell culture techniques
 
Substrates for animal cellculture
Substrates for animal cellcultureSubstrates for animal cellculture
Substrates for animal cellculture
 
Cell lines seminar
Cell lines seminarCell lines seminar
Cell lines seminar
 
Growth and maintenance of cells
Growth and maintenance of cellsGrowth and maintenance of cells
Growth and maintenance of cells
 

Viewers also liked

Building Historical Geographical Datasets with DBpedia
Building Historical Geographical Datasets with DBpediaBuilding Historical Geographical Datasets with DBpedia
Building Historical Geographical Datasets with DBpediaodhrangavin
 
Florida Supercon 2016 Program Guide
Florida Supercon 2016 Program GuideFlorida Supercon 2016 Program Guide
Florida Supercon 2016 Program GuideSandy Martin
 
RESUME AS MODIFIED 21 Apr 2014
RESUME AS MODIFIED 21 Apr 2014RESUME AS MODIFIED 21 Apr 2014
RESUME AS MODIFIED 21 Apr 2014Jim Prendergast
 
Finalised combined booklet
Finalised combined bookletFinalised combined booklet
Finalised combined bookletAvinash SINGHAL
 
Instalacion de sistemas operativos windows y linux
Instalacion de sistemas operativos windows y linuxInstalacion de sistemas operativos windows y linux
Instalacion de sistemas operativos windows y linuxsupernova2015
 
CV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURIN
CV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURINCV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURIN
CV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURINPierre-David Sabourin
 

Viewers also liked (14)

Building Historical Geographical Datasets with DBpedia
Building Historical Geographical Datasets with DBpediaBuilding Historical Geographical Datasets with DBpedia
Building Historical Geographical Datasets with DBpedia
 
Florida Supercon 2016 Program Guide
Florida Supercon 2016 Program GuideFlorida Supercon 2016 Program Guide
Florida Supercon 2016 Program Guide
 
RESUME AS MODIFIED 21 Apr 2014
RESUME AS MODIFIED 21 Apr 2014RESUME AS MODIFIED 21 Apr 2014
RESUME AS MODIFIED 21 Apr 2014
 
Epistaxis
EpistaxisEpistaxis
Epistaxis
 
m2006 staging
m2006 stagingm2006 staging
m2006 staging
 
Finalised combined booklet
Finalised combined bookletFinalised combined booklet
Finalised combined booklet
 
Instalacion de sistemas operativos windows y linux
Instalacion de sistemas operativos windows y linuxInstalacion de sistemas operativos windows y linux
Instalacion de sistemas operativos windows y linux
 
Notification main june2015
Notification main june2015Notification main june2015
Notification main june2015
 
Certificate.
Certificate.Certificate.
Certificate.
 
Final Draft Aug 29 2014
Final Draft Aug 29 2014Final Draft Aug 29 2014
Final Draft Aug 29 2014
 
CV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURIN
CV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURINCV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURIN
CV_ENG_19_FEB_2015_PIERRE-DAVID_SABOURIN
 
ATPER_Krisna Presentation_2016
ATPER_Krisna Presentation_2016ATPER_Krisna Presentation_2016
ATPER_Krisna Presentation_2016
 
pcl cv
pcl cvpcl cv
pcl cv
 
Aundrea M. Travis
Aundrea M. TravisAundrea M. Travis
Aundrea M. Travis
 

Similar to Human Prenatal Small Intestine Cells as a Valuable Source of Stem Cells and Epithelial Cells: Phenotypic and Functional Characterization

#1 principles of tissue engineering
#1 principles of tissue engineering#1 principles of tissue engineering
#1 principles of tissue engineeringRoshan Shetty
 
Articles of scaffold of our research in CTC
Articles of scaffold of our research in CTCArticles of scaffold of our research in CTC
Articles of scaffold of our research in CTCMaisa AlZghoul
 
Histogenic Study of Human Foetal Endocrine Pancreas
Histogenic Study of Human Foetal Endocrine PancreasHistogenic Study of Human Foetal Endocrine Pancreas
Histogenic Study of Human Foetal Endocrine Pancreasiosrjce
 
Cellular totipotency in plants
Cellular totipotency in plantsCellular totipotency in plants
Cellular totipotency in plantsRicha Khatiwada
 
Respiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestineRespiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestineAndrew Fabich
 
Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...
Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...
Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...cordbloodsymposium
 
basic of animal cell culture
basic of animal cell culturebasic of animal cell culture
basic of animal cell cultureashutosh mahale
 
Animal Cells As Host
Animal Cells As HostAnimal Cells As Host
Animal Cells As HostNat Rice
 
Human placental explants in culture approaches and assessments
Human placental explants in culture approaches and assessmentsHuman placental explants in culture approaches and assessments
Human placental explants in culture approaches and assessmentsdjrivelli
 
256447 stem-cell-transplantation
256447 stem-cell-transplantation256447 stem-cell-transplantation
256447 stem-cell-transplantationYoAmoNYC
 
256447 stem-cell-transplantation
256447 stem-cell-transplantation256447 stem-cell-transplantation
256447 stem-cell-transplantationabctutor
 
The Awesome Science of Animal Cell Culture...
The Awesome Science of Animal Cell Culture...The Awesome Science of Animal Cell Culture...
The Awesome Science of Animal Cell Culture...Alok Bharadwaj N
 
Caenorhabditi Elegans Research Paper
Caenorhabditi Elegans Research PaperCaenorhabditi Elegans Research Paper
Caenorhabditi Elegans Research PaperLaura Benitez
 
Applications of cell lines
Applications of cell linesApplications of cell lines
Applications of cell linesKAUSHAL SAHU
 
Martin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicineMartin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicineigorod
 
De novo kidney regeneration with stem cells
De novo kidney regeneration with stem cellsDe novo kidney regeneration with stem cells
De novo kidney regeneration with stem cellsSheila Palacios
 
Advance Access publication 29 January, 2004Establishment o.docx
Advance Access publication 29 January, 2004Establishment o.docxAdvance Access publication 29 January, 2004Establishment o.docx
Advance Access publication 29 January, 2004Establishment o.docxgalerussel59292
 

Similar to Human Prenatal Small Intestine Cells as a Valuable Source of Stem Cells and Epithelial Cells: Phenotypic and Functional Characterization (20)

International Journal of Stem Cells & Research
International Journal of Stem Cells & ResearchInternational Journal of Stem Cells & Research
International Journal of Stem Cells & Research
 
#1 principles of tissue engineering
#1 principles of tissue engineering#1 principles of tissue engineering
#1 principles of tissue engineering
 
Articles of scaffold of our research in CTC
Articles of scaffold of our research in CTCArticles of scaffold of our research in CTC
Articles of scaffold of our research in CTC
 
Histogenic Study of Human Foetal Endocrine Pancreas
Histogenic Study of Human Foetal Endocrine PancreasHistogenic Study of Human Foetal Endocrine Pancreas
Histogenic Study of Human Foetal Endocrine Pancreas
 
Cellular totipotency in plants
Cellular totipotency in plantsCellular totipotency in plants
Cellular totipotency in plants
 
Respiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestineRespiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestine
 
Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...
Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...
Deriving Mesenchymal Stem Cells from Human Amniotic Fluid – Potential for an ...
 
basic of animal cell culture
basic of animal cell culturebasic of animal cell culture
basic of animal cell culture
 
Cell culture
Cell culture Cell culture
Cell culture
 
Animal Cells As Host
Animal Cells As HostAnimal Cells As Host
Animal Cells As Host
 
Human placental explants in culture approaches and assessments
Human placental explants in culture approaches and assessmentsHuman placental explants in culture approaches and assessments
Human placental explants in culture approaches and assessments
 
256447 stem-cell-transplantation
256447 stem-cell-transplantation256447 stem-cell-transplantation
256447 stem-cell-transplantation
 
256447 stem-cell-transplantation
256447 stem-cell-transplantation256447 stem-cell-transplantation
256447 stem-cell-transplantation
 
The Awesome Science of Animal Cell Culture...
The Awesome Science of Animal Cell Culture...The Awesome Science of Animal Cell Culture...
The Awesome Science of Animal Cell Culture...
 
Caenorhabditi Elegans Research Paper
Caenorhabditi Elegans Research PaperCaenorhabditi Elegans Research Paper
Caenorhabditi Elegans Research Paper
 
Applications of cell lines
Applications of cell linesApplications of cell lines
Applications of cell lines
 
Animal cell culture (Basics)
Animal cell culture (Basics)Animal cell culture (Basics)
Animal cell culture (Basics)
 
Martin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicineMartin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicine
 
De novo kidney regeneration with stem cells
De novo kidney regeneration with stem cellsDe novo kidney regeneration with stem cells
De novo kidney regeneration with stem cells
 
Advance Access publication 29 January, 2004Establishment o.docx
Advance Access publication 29 January, 2004Establishment o.docxAdvance Access publication 29 January, 2004Establishment o.docx
Advance Access publication 29 January, 2004Establishment o.docx
 

More from ◂ Justin (M) Gaines ▸

More from ◂ Justin (M) Gaines ▸ (13)

Thermo Fisher 2015 Innovation Day Semifinalist
Thermo Fisher 2015 Innovation Day SemifinalistThermo Fisher 2015 Innovation Day Semifinalist
Thermo Fisher 2015 Innovation Day Semifinalist
 
DV Biologics at a glance...
DV Biologics at a glance...DV Biologics at a glance...
DV Biologics at a glance...
 
DV Biologics Product Menu
DV Biologics Product MenuDV Biologics Product Menu
DV Biologics Product Menu
 
Chronic Kidney Disease & Human Renal Fibroblasts
Chronic Kidney Disease & Human Renal FibroblastsChronic Kidney Disease & Human Renal Fibroblasts
Chronic Kidney Disease & Human Renal Fibroblasts
 
Cell_Lines_HTS_HCS
Cell_Lines_HTS_HCSCell_Lines_HTS_HCS
Cell_Lines_HTS_HCS
 
Primary_Cells_Recombinant_Cell_Lines
Primary_Cells_Recombinant_Cell_LinesPrimary_Cells_Recombinant_Cell_Lines
Primary_Cells_Recombinant_Cell_Lines
 
Biosensors_multiplexed_screening_890_compounds
Biosensors_multiplexed_screening_890_compoundsBiosensors_multiplexed_screening_890_compounds
Biosensors_multiplexed_screening_890_compounds
 
Administration of Autologous Bone Marrow Stem Cells Into Spinal Cord Injury P...
Administration of Autologous Bone Marrow Stem Cells Into Spinal Cord Injury P...Administration of Autologous Bone Marrow Stem Cells Into Spinal Cord Injury P...
Administration of Autologous Bone Marrow Stem Cells Into Spinal Cord Injury P...
 
DV Biologics Overview
DV Biologics OverviewDV Biologics Overview
DV Biologics Overview
 
Keratinocyte selection plan
Keratinocyte selection planKeratinocyte selection plan
Keratinocyte selection plan
 
Human Primary and Cultured Cells (Toxicology Spotlight)
Human Primary and Cultured Cells (Toxicology Spotlight)Human Primary and Cultured Cells (Toxicology Spotlight)
Human Primary and Cultured Cells (Toxicology Spotlight)
 
Nomad Biosensors for HCS
Nomad Biosensors for HCSNomad Biosensors for HCS
Nomad Biosensors for HCS
 
DV Biologics_Brochure
DV Biologics_BrochureDV Biologics_Brochure
DV Biologics_Brochure
 

Recently uploaded

9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 60009654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000Sapana Sha
 
Pulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceuticsPulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceuticssakshisoni2385
 
Labelling Requirements and Label Claims for Dietary Supplements and Recommend...
Labelling Requirements and Label Claims for Dietary Supplements and Recommend...Labelling Requirements and Label Claims for Dietary Supplements and Recommend...
Labelling Requirements and Label Claims for Dietary Supplements and Recommend...Lokesh Kothari
 
Biopesticide (2).pptx .This slides helps to know the different types of biop...
Biopesticide (2).pptx  .This slides helps to know the different types of biop...Biopesticide (2).pptx  .This slides helps to know the different types of biop...
Biopesticide (2).pptx .This slides helps to know the different types of biop...RohitNehra6
 
Disentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTDisentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTSérgio Sacani
 
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bNightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bSérgio Sacani
 
Formation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksFormation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksSérgio Sacani
 
GBSN - Biochemistry (Unit 1)
GBSN - Biochemistry (Unit 1)GBSN - Biochemistry (Unit 1)
GBSN - Biochemistry (Unit 1)Areesha Ahmad
 
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...anilsa9823
 
Pests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdfPests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdfPirithiRaju
 
Zoology 4th semester series (krishna).pdf
Zoology 4th semester series (krishna).pdfZoology 4th semester series (krishna).pdf
Zoology 4th semester series (krishna).pdfSumit Kumar yadav
 
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.Nitya salvi
 
Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsAArockiyaNisha
 
Presentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptxPresentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptxgindu3009
 
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...ssifa0344
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Sérgio Sacani
 
VIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C PVIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C PPRINCE C P
 
Biological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfBiological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfmuntazimhurra
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )aarthirajkumar25
 

Recently uploaded (20)

9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 60009654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
 
Pulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceuticsPulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceutics
 
Labelling Requirements and Label Claims for Dietary Supplements and Recommend...
Labelling Requirements and Label Claims for Dietary Supplements and Recommend...Labelling Requirements and Label Claims for Dietary Supplements and Recommend...
Labelling Requirements and Label Claims for Dietary Supplements and Recommend...
 
Biopesticide (2).pptx .This slides helps to know the different types of biop...
Biopesticide (2).pptx  .This slides helps to know the different types of biop...Biopesticide (2).pptx  .This slides helps to know the different types of biop...
Biopesticide (2).pptx .This slides helps to know the different types of biop...
 
Disentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTDisentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOST
 
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bNightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
 
Formation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksFormation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disks
 
GBSN - Biochemistry (Unit 1)
GBSN - Biochemistry (Unit 1)GBSN - Biochemistry (Unit 1)
GBSN - Biochemistry (Unit 1)
 
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
 
Pests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdfPests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdf
 
Zoology 4th semester series (krishna).pdf
Zoology 4th semester series (krishna).pdfZoology 4th semester series (krishna).pdf
Zoology 4th semester series (krishna).pdf
 
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
 
Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based Nanomaterials
 
The Philosophy of Science
The Philosophy of ScienceThe Philosophy of Science
The Philosophy of Science
 
Presentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptxPresentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptx
 
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
 
VIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C PVIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C P
 
Biological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfBiological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdf
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )
 

Human Prenatal Small Intestine Cells as a Valuable Source of Stem Cells and Epithelial Cells: Phenotypic and Functional Characterization

  • 1. 1Cell & Tissue Transplantation & Therapy 2014:6 Open Access: Full open access to this and thousands of other papers at http://www.la-press.com. Cell & Tissue Transplantation & Therapy Human Prenatal Small Intestine Cells as a Valuable Source of Stem Cells and Epithelial Cells: Phenotypic and Functional Characterization Rami Nasrallah, Toai Nguyen, Anasua Kusari, Kelly Burgee and Rafael Gonzalez DaVinci Biosciences LLC, Costa Mesa, California, USA. ABSTRACT: As the fastest self-regenerating organ, the intestinal epithelium serves primarily to absorb nutrients and minerals, while its other distinct functional characteristics include the capacity to proliferate and replicate many lineage precursors. Currently, in vitro studies using small intestine cells are hindered by the lack of a standard cell culture model. Here, we characterize primary human prenatal small intestine epithelial cells (HPIEC). The data demonstrate that HPIEC express markers indicative of stem cells (NOTCH1, SOX9, OLFM4, LGR5), epithelial cells (CK18, CDH1), and immunological and entero-endocrinal cells (DEFA5, GPR-120, GLP-1). More importantly, HPIEC are shown to secrete GLP-1 and lysozyme, and express sucrase–isomaltase, maltase–glucoamylase, and drug transport function that is cyclosporin A repressible, which demonstrates functionality. These data indicate that HPIEC are a heterogeneous cell population that contains stem cells and epithelial cells that may be valuable for various functional, developmental, and pharmacologic studies. KEYWORDS: in vitro, small intestine cells, characterization, stem cells CITATION: Nasrallah et al. Human Prenatal Small Intestine Cells as a Valuable Source of Stem Cells and Epithelial Cells: Phenotypic and Functional Characterization. Cell & Tissue Transplantation & Therapy 2014:6 1–9 doi:10.4137/CTTT.S14354. RECEIVED: January 17, 2014. RESUBMITTED: March 31, 2014. ACCEPTED FOR PUBLICATION: March 31, 2014. ACADEMIC EDITOR: David T. Harris, Editor in Chief TYPE: Original Research FUNDING: This research was supported by DaVinci Biosciences. The authors alone are responsible for the content and writing of the paper. COMPETING INTERESTS: Authors disclose no potential conflicts of interest. COPYRIGHT: © the authors, publisher and licensee Libertas Academica Limited. This is an open-access article distributed under the terms of the Creative Commons CC-BY-NC 3.0 License. CORRESPONDENCE: rgonzalez@dvbiosciences.com Introduction The small intestine plays a crucial role in digestion and ­nutrient absorption, in constant competition with a plethora of microbes and their toxins which are part of the normal intestinal flora. As the physical boundary between the outside and inside environments, the intestinal epithelium is derived from stem cells. These stem cells reside at the base of the crypt and provide all the lineages of epithelial cells, as they con- tinuously divide, differentiate, and migrate toward the apex of the villus from where they slough off.1,2  The epithelium of the adult mammalian intestine is in constant dynamic interaction with its underlying mesenchyme controlling direct progenitor proliferation, lineage commitment, terminal differentiation, and ultimately cell death.2 Small intestine epithelial cells are potentially a suitable model for researching inflammatory bowel disease, ­colitis, intestinal cancer, intestinal infections and more importantly, toxicity and drug discovery. Although recent research has described a variety of culture methods,3–7   no simplified long-term culture system has been established from human prenatal intestinal tissue that maintains basic crypt–villus physiology and metabolic functionality in the differentiated state. The majority of studies are not performed on primary intestinal epithelial cells that have been derived directly from human intestinal tissue. This is in part due to limited access to viable human intestines and the complexity to isolate, purify, and propagate primary human intestinal epithelial cells. As an alternative, commercially available intestinal epithelial cell lines such as Caco-2 and HT29 are used.8–10  Caco-2, which is one of the most widely used and best described intestinal cell models, is derived from a neoplastic tumor of the large intestine of a 72-year-old man.11  Although these cells could be useful for studies on cell growth, the data obtained from studies on proliferation and ion transport do not ­necessarily support the results from the experiments performed with primary human intestinal epithelial cells.12  Since the use of
  • 2. Nasrallah et al 2 Cell & Tissue Transplantation & Therapy 2014:6 these cell lines as tools has limitations,2,5,9,10  efforts have been directed at developing alternative sources of human intesti- nal epithelial cells that maintain their functions and are easy to work with. Although culture systems of human intestinal cells have been described,2–9  an adequate and easily obtain- able primary cell culture model that is characterized and demonstrates functionality does not exist. The ideal primary cell model would contain the complete flora of cells normally found in the small intestine, be reproducible, expandable, and functional in vitro. Here, we present phenotypic, molecular biological, and functional characterization of primary human prenatal small intestine epithelial cells (HPIECs) in vitro. These small intestine cells are a heterogeneous population that is eas- ily expandable, possesses reproducible phenotypic proper- ties and may possibly evade the immune response as is the case in fetal cell microchimerism,13  which would be suitable for allogeneic transplant. HPIEC express stem cell markers, LGR5, SOX9, NOTCH1, and OLFM4;14,15 mature epithe- lial cell marker, CK18; and innate immune marker, DEFA5. HPIEC maintain the dynamic balance between proliferation– differentiation unchanged for several passages. HPIEC also maintain stem cells and terminally differentiated cells. In addition, the data demonstrate that HPIEC inducibly secrete peptide GLP-1  and anti-microbial enzyme lysozyme, and express P-glycoprotein (Pgp) multi-drug transporter, sucrase–­ isomaltase, and maltase–glucoamylase. Collectively, these properties suggest that these cells are a potential in vitro model for drug discovery and development, and possibly in cell transplantation and tissue engineering studies. Materials and Methods Cell culture. HPIEC were isolated from 17- to 18-week- old pre-natal small intestine tissue made available from elec- tive medical abortions, with informed consent of donors. The study and consent procedure were approved by an independent institutional review board (Ethical & Independent Review Services IRB 00007807; Study #08103–05). Cell dissocia- tion solution, culture vessel coating solution, and epithelial pro-conditioned (D-Pro) medium were all from DV Biolog- ics, LLC (Costa Mesa, CA). The epithelial pro-conditioned medium was generated by culturing intestinal mesenchy- mal cells to 80–90% confluency using DMEM high glucose (Gibco, Carlsbad, CA) and 10% fetal bovine serum (FBS; Hyclone, Logan, UT), and then switching the media to low serum, 2% FBS, to stress the cells. The conditioned media were harvested after each medium replacement. These ­conditioned media were then combined 50/50 with basal media ­containing minimum essential medium (Gibco) supplemented with insu- lin/transferrin/selenium (BD Biosciences, San Diego, CA), using 20  ng/ml EGF and 10  ng/ml HGF (Peprotech, Rocky Hill, NJ), 200  ng/ml choleratoxin and 40  µg/ml heparin (Sigma-Aldrich, St Louis, MO) as per manufac- turer’s instruction. All cells used for experiments were tested for authentication using methods described below. Primary HPIEC were seeded (1.5–4 × 104  cells/cm2 ) in vessels (25 cm2 , or 6-well plates) treated with culture vessel coating solution and cultured in epithelial pro-conditioned medium (DV Bio- logics). To remove dead cells and cellular debris, the medium was replaced 24 hours after seeding, so cell clusters could fully adhere to the growth surface. Afterward, medium change was performed every two days. To maximize the number of serial passages, sub-culture was performed when the cells were 80 to 90% confluent (4–8 days). Growth kinetics. HPIEC were seeded in duplicates at 1.5 × 104  to 2.0 × 104  cells/cm2  in 6-well plates treated with culture vessel coating solution in epithelial pro-conditioned medium. The cells were dissociated with cell dissociation solu- tion every seven days, counted, and sub-cultured at the same seeding density. Population doubling was calculated using the formula PD  =  [log10 (N1 )  -  log10 (N0 )]/log10 (2) as previously described.16 Colony forming unit assay. HPIEC were resuspended in epithelial pro-conditioned medium and seeded onto six 10 cm dishes. One dish was seeded with 1.0 × 104  cells (control) and the remaining five dishes were seeded with 1.0 × 102  cells (experimental). The plated cells were incubated in the CO2   incubator for two weeks. Culture dishes were rinsed with PBS, fixed (4% formaldehyde and 1% glutaralde- hyde in PBS), stained with 1% crystal violet in 10% methanol (all from Fisher Scientific, Pittsburgh, PA), destained with water, and air dried. Colonies were quantified using Bio-Rad Quantity One v. 4.6.6  software and imaging system (Bio- Rad, Hercules, CA). Immunocytochemistry. Cells were cultured in 6-well plates, fixed in phosphate-buffered 4% paraformaldehyde (PFA) and stored at 4°C. After permeabilization with 0.1% of Triton X-100 for internal antigens (Promega, Madison, WI) and blocking with 1% BSA (Sigma-Aldrich) and 10% Nor- mal Donkey Serum (Jackson Immunoresearch, West Grove, PA), primary antibodies were applied overnight at 4°C. Cells were incubated with secondary antibody in blocking buffer for one hour at room temperature (RT). Cells were counterstained with DAPI (Molecular Probes, Carlsbad, CA) and mounted with Fluoromount-G (Southern Biotech, Birmingham, AL). Primary antibodies used were mouse anti-E-cadherin, rab- bit anti-Ki67, rabbit anti-ZO1, mouse anti-CK18 (Millipore, Billerica, MA), and rabbit anti-GPCR GPR-120, mouse anti- SOX9, and rabbit anti-GPCR GPR49 (anti-LGR5) (Abcam, Cambridge, England). Secondary antibodies Alexa 488 and Alexa 594  (Molecular Probes, Carlsbad, CA) were used. As negative controls corresponding specific non-immune immunoglobulins (Santa Cruz Biotech, Santa Cruz, CA) were used for all experiments. Staining was analyzed using an Olympus IX81 inverted microscope and SlideBook 4.2 soft- ware (Intelligent Imaging Innovations, Inc.). PCR analysis. RNA was directly isolated from primary or cultured HPIEC and brain tissue using the RNeasy Mini
  • 3. Human prenatal small intestine cells as a valuable source stem and epithelial cells 3Cell & Tissue Transplantation & Therapy 2014:6 Kit (Qiagen, Hilden, Germany). cDNA was synthesized with Superscript III (Invitrogen, Carlsbad, CA) using ran- dom hexamers and 200 ng of total RNA in a 25-µl reaction. PCR was conducted in a C1000TM Thermal Cycler (Bio-Rad, Hercules, CA) using GoTaq® Green Master Mix (Promega) and 2 µl of cDNA product for the analyses of all genes. No RT controls were included in each experiment. RNA from brain tissue was used as a control to show expression of these genes in another tissue type. PCR products were visualized by aga- rose gel electrophoresis. Table 1 lists PCR primer sequences used for all experiments. Lysozyme functional assay. The lysozyme assay was performed according to manufacturer’s protocol (Worthing- ton Biochemical, Lakewood, NJ) with modifications. Briefly, HPIEC were incubated in triplicates for two hours in assay buffer alone DMEM high glucose (Invitrogen) plus 15% FBS (HyClone) and as applicable supplemented with 0.3  mg/ ml Micrococcus lysodeikticus. The clarified supernatants were assayed for their ability to lyse a suspension of M. lysodeikticus over time, by recording the absorbance at 450 nm every four ­minute; a solution of 5 U/ml purified lysozyme (Worthing- ton Biochemical) in assay buffer was processed in parallel as a control. GLP-1  assay. ELISA was performed according to the manufacturer’s suggested protocol for GLP-1 assay kit (Mil- lipore). Briefly, HPIEC were cultured in duplicates in 12-well plates for four days. The cells were washed twice with a rinse buffer (PBS supplemented with CaCl2   and MgCl2 , 2  and 1 mmol/l,respectively,and 0.5% BSA),incubated for two hours at 37°C, 5% CO2 , in 500 µl rinse buffer containing 0.1 mmol/l Diprotin A (Sigma-Aldrich), supplemented with glucose (5 or 25 mmol/l); or with tolbutamide and/or forskolin and isobutyl- methylxanthine (IBMX) (10, 10, and 500 µmol/l, respectively). Tolbutamide, forskolin, and IBMX (all from Sigma-Aldrich) were diluted from 1000× stock solutions in DMSO. Solu- tions with equivalent final DMSO concentrations were used as controls for samples containing tolbutamide, and/or forskolin and IBMX. GLP-1 in clarified supernatants was quantified by ELISA using a VMax ­Microplate Reader (Molecular Devices, Sunnyvale, CA). Multidrug transporter assay. The experiment was per- formedusingVybrant®MultidrugResistanceAssayKitaccord- ing to the manufacturer’s suggested protocol (Invitrogen). Briefly, HPIEC and Caco-2 cells were incubated in growth medium (DMEM supplemented with 15% FBS) with 0, 0.165, 1.48, 13.3, and 120 mg/l cyclosporin A for 15 min, exposed Table 1. Primer sequences used in PCR studies. GENE SEQUENCE AMPLICON SIZE CK14 ACGATGGCAAGGTGGTGT 118 bp GGGATCTTCCAGTGGGATCT DEFA5 TGAGGACCATCGCCATCCTTGCT 226 bp TCACGGGTAGCACAACGGCCG GAPDH CAAGGTCATCCATGACAACTTTG 496 bp GTCCACCACCCTGTTGCTGTAG OLFM4 CCAGCTAAGAGGACAAGATGAG 286 bp GGCAGGGAAACAGAGCAC LGR5 GAACATGCTCACGGGAGTCTC 303 bp CCACCCAGCAGGGGAAC CK18 GGGAGCACTTGGAGAAGAAG 229 bp GATATTGGTGTCATCAATGACCT E-cadherin-1 AGGAACACAGGAGTCATCAGTG 260 bp GGGGTATTGGGGGCATC GRP120 GCTCTGCCTCTCTGCGTCT 255 bp GCGGATCTGGTGGCTCTC GLP1 ATCATTCTCAGCTTCCCAGG 370 bp CTCATCAGAGAAAGAACCATCAG NOTCH-1 CGCCTTTGTGCTTCTGTTC 300 bp GGTGGTCTGTCTGGTCGTC Sucrase Isomaltase TTCGCTACACCTTATTACCCTTCC 415 bp TCTCCTTTGGCTGTGTTGTTTTC Maltase Glucoamylase CCCTTCGCCTGGATGTCAC 371 bp TGGGGGCTGGTCTCGG
  • 4. Nasrallah et al 4 Cell & Tissue Transplantation & Therapy 2014:6 to 0.25 µmol/l calcein acetoxymethyl ester (calcein-AM) for 15 min, washed twice with growth medium, and the fluores- cence was quantified using the Bio-Rad CFX-96 RT system. The results were normalized to no cyclosporin A control. Results Propagation of HPIEC. Since there is a current absence of a small intestine primary cell model, we took on the task to characterize small intestine cells isolated from prenatal ­tissue and propagate them using a conditioned media obtained from intestine-derived mesenchymal cells. The culture sys- tem described herein has been optimized specifically for the culture of small intestinal epithelial cells as multi-cellular aggregates (organoids), since previous experimentation with intestinal epithelial cells indicated that breaking down the epithelium to a single-cell suspension led to poor adherence and survival of the epithelial cells. Initially seeded as organ- oids (Fig. 1A), HPIEC exhibit better long-term viability in culture in comparison to starting from a single-cell suspension (personal observation, data not shown). In D-PRO medium, these organoids quickly formed a lawn of highly enriched small intestine epithelial cells (Fig. 1B). Easily sub-cultured with the use of cell dissociation solution, HPIEC maintained their phenotypical morphology after four passages or nine population doublings (Fig. 1C). HPIEC could be propagated for at least six passages or 14 population doublings without any noticeable morphological changes (Fig. 1D). HPIEC characterization. To better describe HPIEC, we performed molecular and immunological characteriza- tions using immunocytochemistry (ICC) and RT-PCR. ICC analysis demonstrates that HPIEC express division and ­proliferative marker Ki67 (Fig. 2A). Similar to Ki67 ICC stain- ing, the stem cell marker SOX9 is also expressed in some of the HPIEC (Fig. 2A). This may explain why HPIEC are eas- ily expandable. The staining of both Ki67 and SOX9 appears to be confined to the inner cells of epithelial colonies that are formed. Interestingly, within organoids, the epithelial stem cell marker LGR-5 is also found (Fig. 2B). Our RT-PCR data support and confirm our ICC results. PCR analysis demon- strates that HPIEC express early development and stem cell markers NOTCH1, SOX9, OLFM4, and LGR5 (Fig. 2C). 0 10 20 30 40 0.0 3.0 6.0 9.0 12.0 15.0 Populationdoubling Days in culture D Figure 1. Growth characteristics of HPIEC. A) Phase contrast of freshly isolated HPIEC. After overnight attachment HPIEC show characteristic organoid structure of intestinal epithelial cells. B) Established culture of HPIEC, Passage 0 phase contrast. After 7 days in culture in epithelial pro conditioned medium, HPIEC display typical morphology of intestinal epithelial cells. C) Established culture of HPIEC, Passage 4 phase contrast. After over 8 population doublings in culture using epithelial pro conditioned medium, HPIEC maintain typical morphology of intestinal epithelial cells. D) HPIEC growth curve illustrating cumulative population doublings.
  • 5. Human prenatal small intestine cells as a valuable source stem and epithelial cells 5Cell & Tissue Transplantation & Therapy 2014:6 In addition, we performed colony forming unit (CFU) assays to confirm the presence of stem cells. When HPIEC were seeded at 100  cells per 10  cm dish, the data illus- trate the formation of 9.6  ±  0.7  CFUs (n  =  5). Control that was seeded at 10,000 cells per 10 cm dish produced a lawn of cells at two weeks (data not shown). HPIEC express markers indicative of mature epithelial cells. HPIEC express adhesion of epithelial cell marker E-cad- herin, tight junction associated protein marker ZO-1, the mature epithelial marker CK18, and intestinal marker G protein coupled receptor 120 (GPR-120) (Fig. 3A). Our PCR data (Fig. 3B) support and extend the ICC results. HPIEC express mature epithelial cell markers CK18 and E-cadherin, and also express the Paneth cell marker defensin-α-5 (DEFA5), which is thought to be involved in host defense. In addition, the markers GPR-120 and glu- cagon-like peptide 1 (GLP-1), which are thought to play an important role in regulating insulin, are also expressed (Fig. 3B). Indeed, our PCR analysis of epithelial cell markers (CK-18 and CDH1), epithelial stem cells markers (LGR5, SOX9, and OLFM4), and Paneth cells (DEFA5) demonstrates that cultured cells in conditioned medium maintain the same gene expression profile as uncultured freshly isolated cells (Figs. 2 and 3, lane 2). These data sup- port the notion that HPIEC contain a mixed population of stem cells and terminally differentiated cells. HPIECfunctionality.We observed that HPIEC secrete GLP-1  and lysozyme upon stimulation and express a repressible multidrug transporter function (Fig. 4). Using ­enzyme-linked immunosorbent assay (ELISA), we found that HPIEC secrete GLP-1, a protein synthesized only in the intestine by posttranslational processing of proglucagon via the L-cell.17  HPIEC secrete GLP-1 in response to glucose in a dose-dependent manner and upon stimulation with the anti-diabetes compound tolbutamide and elevated cAMP lev- els that are induced by forskolin and IBMX (Fig. 4A). Paneth cells secrete lysozyme and α-defensins, both of which have clear antimicrobial activity, and also tumor necro- sis factor-alpha, and phospholipase A2.18  For further func- tional testing,HPIEC were exposed to M. Lysodeikticus,which stimulated the secretion of lysozyme (Fig. 4B), thus confirm- ing that the present culture system preserves the subpopula- tion diversity comprising Paneth cells, among others that exists in the natural environment of the small intestine. We also researched for the presence of Pgp multi-drug transporter activity in HPIEC. This was determined using the Invitrogen Vybrant MDR assay kit. This assay kit relies on the passive diffusion of the lipophilic calcein-AM, which can be carried outside the cell by a Pgp multi-drug transporter, if it is not converted to fluorescent calcein by indigenous esterases. The Pgp multi-drug transporter can be inhibited by cyclosporin A, resulting in an intracellular accumulation of fluorescent calcein. The data demonstrate that, similar to Caco-2 cells, in the present culture system, HPIEC express a Pgp ­multi-drug transporter activity, which is inhibited by cyclosporin A in a dose-dependent manner (Fig.4C).Our PCR data ­demonstrate A B C 1 2 3 4 NOTCH1 SOX9 OLFM4 LGR5 GAPDH Figure 2. Immunocytochemical staining and Molecular marker characteristics of HPIEC. HPIEC were stained with specific monoclonal or polyclonal antibodies, counter-stained with DAPI (Blue), and visualized with fluorescence microscopy. Panel A illustrates staining of HPIEC with antibodies specific for SOX9 (green) and Ki67 (red). Merged images demonstrate that not all cells express SOX9 and Ki67. Panel B illustrates staining of LGR5 which is mostly confined to organoid structures. For panel C total RNA was isolated and reverse transcribed (RT); the resulting cDNA was PCR- amplified with primer pairs specific for markers of stem cells NOTCH1, SOX9, OLFM4, and LGR5. GAPDH was included as housekeeping marker control. Lane 1: No RT control; lane 2: Uncultured HPIEC; lane 3: Passage 4 HPIECs; lane 4: Brain tissue.
  • 6. Nasrallah et al 6 Cell & Tissue Transplantation & Therapy 2014:6 that HPIEC express sucrase–isomaltase and ­maltase–glu- coamylase (Fig. 4D).The expression of specific enzyme mark- ers sucrase–isomaltase and maltase–­glucoamylase is ­indicative of the presence of functional entrocytes. ­Moreover, our PCR analysis demonstrates that cultured cells in conditioned medium maintain the same expression profile as uncultured freshly isolated cells (Figs. 4D, lane 2  and  3). These results further support the notion that HPIEC are functional and a valuable source for various studies. Discussion Recent advances in small intestine research illustrate the intimate interaction between intestinal stem cells and the self-renewing epithelium. This interaction can be explained by an intricate interplay among a distinct set of growth sig- nals that are now becoming well defined.15,19  Presently, there is no standard in vitro cell culture system for small intes- tine epithelial cells although several have been reported in the literature.5–7,19  Sato et al described a small intestine cell culture system that is in a 3D matrix and requires organoid formation and maintenance. Although this system may have limitless potential, it does not appear to be a simplified in vitro model and to the best of our knowledge they have yet to test the ability of this model for specific in vitro studies such as GLP-1  secretion and multi-drug transporter assays in human cells.19 The purpose of this study was to identify a simple cell culture system of small intestine epithelial cells that mimics the in vivo system. The system must be easy to work with and should be valuable for multiple in vitro studies. After researching current literature, we decided to use cells isolated from prenatal small intestine. HPIEC are a hetero- geneous monolayer cell population that contains all the cell populations of the small intestine. Monolayer of cells is widely used throughout industry and enables rapid research and drug discovery in vitro. HPIEC exhibit stem cell self-renewal and multipotential differentiation in culture. In addition, the close proximity of Paneth cells to stem cell populations provides intestinal epithelial cells with essential signals to assemble into self-organizing structures. Thus, it is imperative to take into consideration isolation techniques that preserve close cel- lular interactions.3 The culture vessel coating solution and conditioned media of the presently described culture system provides an adequate environment supporting the growth of HPIEC. Current evidence illustrates the significant roles of the extra- cellular matrix (ECM) as well as cell culture media on the attachment, differentiation, and proliferation of intestinal epithelia.3,4,20–22  Since the proliferation of epithelial crypt is Wnt dependent,23,24  the method employed herein also simu- lates the extracellular signals found in the intestinal crypt, by utilizing a medium conditioned by mesenchymal and Paneth cells that reside in the crypt–villus niche. These cells secrete various cytokines that modulate the Wnt gene expression pathway similar to that expressed in vivo.23,24 Although the growth, maintenance, and propagation of small intestinal epithelial cells can be challenging, HPIEC are easily expandable and maintain their phenotype (Fig. 1), most likely due to the method of isolating ­organoids consisting of intact villi and crypt units that have a much greater capacity to adhere, survive, and maintain the polarity of epithelium.3  Base columnar cells in select populations of SOX9 expressing cells in crypt cells were shown to produce multilineage ­intestinal organoids in ex vivo culture systems.25   Indeed, HPIEC express dividing cell marker Ki67  and stem cell markers A B 1 2 3 4 CK18 CDH1 (E-cadherin) DEFA5 GPR-120 GLP-1 GAPDH Figure 3. Immunocytochemical staining and Molecular marker characteristics of HPIEC. HPIEC were stained with specific monoclonal or polyclonal antibodies, counter-stained with DAPI (Blue), and visualized with fluorescence microscopy. Panel A illustrates antibodies specific for E-cadherin (green), ZO-1 (red), cytokeratin-18 (CK18; green) and G protein-coupled receptor 120 (GPR-120; red). For panel B total RNA was isolated and reverse transcribed (RT); the resulting cDNA was PCR-amplified with primer pairs specific for epithelial cells (CK18, CDH1), and entero-immunological and entero-endocrinal characteristics (DEFA5, GPR-120, GLP-1); GAPDH was included as housekeeping marker control. Lane 1: No RT control; lane 2: Uncultured HPIEC; lane 3: Passage 4 HPIECs; lane 4: Brain tissue.
  • 7. Human prenatal small intestine cells as a valuable source stem and epithelial cells 7Cell & Tissue Transplantation & Therapy 2014:6 * * * Control Tolbutamide [GLP-1]inmedium(pM) Forskolin+IBMX Tolb+Forsk+IBMX Glucose5mM Glucose25mM 0 20 40 60 A A450 Elapsed time (min) 0.00 0.20 0.40 0.60 0.80 1.00 0 5 10 15 20 25 B * ** * * * 0.000 0.165 1.481 13.333 80 120 160 200 240 %retention µg/ml Cyclosporin A C D 1 2 3 4 Sucrase-isomaltase Maltase-glucoamylase GAPDH Figure 4. Functional characterization of HPIEC. A) Synthesis of glucagon-like peptide 1 (GLP-1). PD015-F cells were equilibrated in assay buffer for 2 h, then exposed to tolbutamide, or forskolin and isobutylmethylxanthine (IBMX), or tolbutamide, forskolin and IBMX, or 5 mM glucose, or 25 mM glucose as indicated for 2 h. GLP-1 in the clarified supernatants was assayed by ELISA, and normalized to no treatment control. B) Synthesis of lysozyme. PD015-F cells were incubated in assay buffer alone (¡), or challenged with 0.3 mg/ml Micrococcus lysodeikticus (ML; ¨) for 2 h. The ability of the clarified supernatants to lyse a suspension of ML over time was assayed; a solution of 5 U/ml purified lysozyme served as positive control (∆). C) Inhibition of P-glycoprotein activity. PD015-F cells (Lighter shade) and Caco-2 cells (Darker shade) were treated in various concentrations of cyclosporin A for 15 min, and loaded with calcein-AM (0.25 µmol/l) for 15 min; after two washes, and residual fluorescence was quantified, and normalized to no cyclosporin A control. D) Expression of sucrase-isomaltase and maltase-glucoamylase. Total RNA was isolated and reverse transcribed (RT); the resulting cDNA was PCR-amplified with primer pairs specific for sucrase-isomaltase and maltase-glucoamylase. Lane 1: No RT control; lane 2: Uncultured HPIEC; lane 3: Passage 4 HPIECs; lane 4: Brain tissue. Error bar: ± SEM. Asterix (*) denotes statistically significant difference over control. SOX9, NOTCH1, OLFM4 and LGR5 (Fig. 2). Colony form- ing unit assay of HPIEC suggest that approximately 9.6% of the cells may be of the stem cell origin (data not shown). The co-localization of SOX9 and the proliferation marker Ki67 in cultured cells (Fig. 2) illustrate the possibility of using human fetal cells to isolate selective stem populations capable of gen- erating complete villi and crypt in vitro for tissue engineering purposes. Figure 2 also demonstrates the expression of LGR5, a Wnt targeting gene that is selectively expressed in the base of the intestinal crypt. LGR5+ stem cells are critical regula- tors of the cell populations at the crypt base. LGR5+ cells are unique due to their capability to form self-renewing, and mul- tipotent organoids that can recapitulate the structure and cel- lular organization of intestinal epithelium from a single cell.19 HPIEC are a monolayer of cells. They consist of a het- erogeneous population of cells found in the small intestine (Figs. 1–3). HPIEC express markers indicative of mature epi- thelial cells (E-cadherin, ZO-1, CK18; Fig. 3), and immuno- logical and entero-endocrinal cell markers (DEFA5, GPR-120, GLP-1; Fig. 3). These data provide evidence of a natural diver- sity of native subpopulations of ­progenitor cells, stem cells, and their progeny of transit-amplifying cells.2,3,16   HPIEC take advantage of normal stem cell properties related in organoids.19  Future studies will aim to identify percentages of specific cell types within the HPIEC population. Several commercially available intestinal epithelial cell lines are being used for functional studies. These commer- cially available cell lines are derived from transformed human
  • 8. Nasrallah et al 8 Cell & Tissue Transplantation & Therapy 2014:6 ­tissue such as polyps and cancerous lesions (HT-29, Caco- 2, and T84),8–10   which have their limitations that include their malignant nature and colonic origin.5,8   HPIEC are a monolayer cell system that may be similar to native small intestine, which is confirmed by the expression of specific intestinal metabolite enzymes and transporter proteins such as ATP-binding cassette (ABC) transporters Pgp expressed by absorptive enterocytes. Since HPIEC maintain mature and specialized intestinal cell populations after isolation and prop- agation, these cells provide researchers with a tool to inves- tigate digestive and absorptive capacities of normal human intestines (Fig. 4). HPIEC have the ability to secrete GLP-1 upon stimula- tion with glucose or anti-diabetic and cAMP-elevating com- pounds (Fig. 4). GLP-1 is a secretory peptide of physiological importance. It acts as a potent incretin, specifically stimulat- ing glucose-induced insulin release, inhibiting glucagon secre- tion, and modulating the ileal control machinery by slowing down gastric emptying.17,26  As a major regulator of the insu- lin secretory pathway, GLP-1  has many vital physiologi- cal actions, with many great implications in recent diabetes research.16  In addition, it has been shown to affect the central nervous system, with its neuroprotective properties and influ- ence on learning and memory.26  Another important feature of GLP-1 is that it plays an inhibitory role in the regulation of eating and drinking.27  GLP-1 or a potent, long-acting natu- ral or synthetic GLP-1  agonist, such as Exenatide (Byetta) or Liraglutide, were demonstrated to lower glycemia which has resulted in its acceptance as a treatment for patients with type 2 diabetes.28  These data suggest that HPIEC may be a potential research model in the areas of gastrointestinal and neurological disorders, and diabetes therapy. HPIECsecretelysozymeuponexposuretoM.lysodeikticus (Fig. 4) which illustrates that these cells preserve Paneth cell functional properties in the context of the intestinal epithe- lium. Paneth cells, which are characterized by their large eosinophilic granules of anti-microbial compounds, comprise another important subpopulation of the intestinal epithelium. They are located near the crypts that secrete, among other substances, defensin-α and lysozyme, following interaction with bacterial antigens and in response to pharmacologic stimulation.18,29,30  It has been shown that there is decreased expression of α-defensins in Crohn’s iletis.31  Since HPIEC contain Paneth cells, this may serve as a model for research in intestinal immunity, encompassing inflammatory bowel dis- ease, Crohn’s disease, and other ­gastrointestinal disorders.31 Shown to express a Pgp-like function, repressible by cyclosporin A, using calcein-AM as the test substrate, HPIEC may be a suitable model for rapid screening of intesti- nal drug absorption and the detection of chemical compounds ­interfering with Pgp-like functions (Fig. 4C). Intestinal epi- thelium has the ability to transport toxic substances, such as chemotherapy drugs, which may have been taken up non- specifically by passive diffusion or in any other way into the lumen. In some cancers, these efflux functions are highly amplified and contribute to their resistance to chemotherapy. The intestinal epithelial native non-specific efflux function is a current subject in the screening process for suitable anti-cancer drugs that can circumvent the potential problem of resistance. Pgp, the most widely studied ABC transporter, functions as a biological barrier by regulating the cellular accumula- tion and disposition of cytotoxic and xenobiotics drugs out of cells.32,33  In vitro and in vivo studies have demonstrated that Pgp is involved in the export of a variety of natural drugs and lies within the luminal membranes of the endothelial cells of the brain and testes, in the adrenal glands, renal proximal tubule cells, and in the ­apical membrane of epithelial cells of the liver, colon, and small intestine. Responses to external stimuli have been poorly studied in most in vitro culture methods; however, the data demonstrated here show that HPIEC have the indicative feature to respond to stimuli in vitro. The expression of specific enzyme markers sucrase–isomaltase and maltase–glucoamylase is indicative of the presence of functional enterocytes (Fig. 4D). Enterocytes are responsible for many cytokines secretion, inflammatory response, and play a role in microflora adhesion proteins as well as bacterial pathogenesis.11  Enterocytes play a key role in responses to external stimuli and interaction with the gut’s bacterial flora. Thus, cytokine expressing cells are important when considering assembly of responsive architectural model of intestinal tissue. In summary, here we have characterized a prenatal, het- erogeneous primary small intestine epithelial cell which we designate HPIEC, which express several stem cell and mature epithelial cell markers. More importantly, the data illus- trate that HPIEC may be an appropriate and adequate cell source for in vitro research in gastrointestinal disorders, dia- betes research, drug discovery and tissue engineering studies since they contain stem cells (LGR5+), enterocytes (sucrase–­ isomaltase and maltase–glucoamylase expression), Paneth cells (lysozyme secretion), and enteroendocrine cells (GLP-1 secre- tion). Future studies will aim to quantify specific cell types in HPIEC and target growing the cells in a 3D environment or using biodegradable matrices in order to reconstitute the small intestine environment. Such studies precipitate research to help individuals suffering from intestinal cancers and pos- sibly deficiencies of sucrase–isomaltase. Acknowledgements Authors are thankful to Dr. Nickolas Chelyapov and Dr. Robert Sackstein for manuscript review and suggestions. Author Contributions Conceived and designed the experiments: RN, TN, RG. Analyzed the data: RN, TN, RG. Wrote the first draft of the manuscript: RN, RG. Contributed to the writing of the ­manuscript: TN, AK, KB. Agree with manuscript results and conclusions: RN, TN, AK, KB, RG. Jointly developed
  • 9. Human prenatal small intestine cells as a valuable source stem and epithelial cells 9Cell & Tissue Transplantation & Therapy 2014:6 the structure and arguments for the paper: RN, TN, RG. All authors reviewed and approved of the final manuscript. DISCLOSURES AND ETHICS This paper was subject to independent, expert peer review by a minimum of two blind peer reviewers. All editorial decisions were made by the independent academic edi- tor. All authors have provided signed confirmation of their compliance with ethical and legal obligations including (but not limited to) use of any copyrighted material, com- pliance with ICMJE authorship and competing interests disclosure guidelines and, where applicable, compliance with legal and ethical guidelines on human and animal research participants. REFERENCES 1. Dignass AU, Tsunekawa S, Podolsky DK. Fibroblast growth factors modulate intestinal epithelial cell growth and migration. Gastroenterology. 1994;106: 1254–1262. 2. Chopra DP, Dombkowski AA, Stemmer PM, Parker GC. Intestinal epithelial cells in vitro. Stem Cells Dev. 2010;19:131–142. 3. Evans GS, Flint N, Somers AS, Eyden B, Potten CS. The development of a method for the preparation of rat intestinal epithelial cell primary cultures. J Cell Sci. 1992;101:219–231. 4. Perreault N, Beaulieu J. Primary cultures of fully differentiated and pure human intestinal epithelial cells. Exp Cell Res. 1998;245:34–42. 5. Pageot LP, Perreault N, Basora N, Francoeur C, Magny P, Beaulieu JF. Human cell models to study small intestinal functions: recapitulation of the crypt-villus axis. Microsc Res Tech. 2000;49:394–406. 6. Liu Z, Zhang P, Zhou Y, Qin H, Shen T. Culture of human intestinal epithelial cell using the dissociating enzyme thermolysin and endothelin-3. Braz J Med Biol Res. 2010;43:451–459. 7. Sato T. Novel intestinal stem cell culture system. Inflamm Regen. 2012;32:43–47. 8. Rousset M. The human colon carcinoma cell lines HT-29  and Caco-2: two in vitro models for the study of intestinal differentiation. Biochimie. 1986;68: 1035–1040. 9. Parlesak A, Haller D, Brinz S, Baeuerlein A, Bode C. Modulation of cytokine release by differentiated CACO-2 cells in a compartmentalized coculture model with mononuclear leucocytes and nonpathogenic bacteria. Scand J Immunol. 2004;60:477–485. 10. Huang DB, DuPont HL, Jiang ZD, Carlin L, Okhuysen PC. Interleukin-8 response in an intestinal HCT-8 cell line infected with enteroaggregative and enterotoxigenic Escherichia coli. Clin Diagn Lab Immunol. 2004;11:548–551. 11. Grajek W, Olejnik A. Epithelial cell cultures in vitro as a model to study func- tional properties of food. Pol J Food Nutr Sci. 2004;1(3/54):5–24. 12. Jung HC, Eckmann L, Yang SK, et al. A distinct array of proinflammatory cyto- kines is expressed in human colon epithelial cells in response to bacterial inva- sion. J Clin Invest. 1995;95:55–65. 13. Bianchi DW, Zickwold GK, Weil GJ, Sylvester S, DeMaria MA. Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc Natl Acad Sci U S A. 1996;93(2):705–708. 14. Formeister EJ, Sionas AL, Lorance DK, Barkley CL, Lee GH, Magness ST. Distinct SOX9  levels differentially mark stem/progenitor populations and enteroendocrine cells of the small intestine epithelium. Am J Physiol Gastrointest Liver Physiol. 2009;296:G1108–G1118. 15. Sato T, van Es JH, Snippert HJ, et al. Paneth cells constitute the niche for LGR5 stem cells in intestinal crypts. Nature. 2011;469:415–418. 16. Cristofalo VJ, Allen RG, Pignolo RJ, Martin BG, Beck JC. Relationship between donor age and the replicative lifespan of human cells in cultura: a reevaluation. Proc Natl Acad Sci U S A. 1998;95:10614–10619. 17. Holst JJ. Enteroglucagon. Annu Rev Physiol. 1997;59:257–271. 18. Peeters T, Vantrappen G. The Paneth cell: a source of intestinal lysozyme. Gut. 1975;16:553–558. 19. Sato T, Vries RG, Snippert HJ, et al. Single LGR5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–266. 20. Goke M, Zuk A, Podolsky DK. Regulation and function of extracellular matrix intestinal epithelial restitution in vitro. Am J Physiol Gastrointest Liver Physiol. 1996;271:G729-G740. 21. Sanderson IR, Ezzell RM, Kedinger M, et al. Human fetal enterocytes in vitro: modulation of the phenotype by extracellular matrix. Proc Natl Acad Sci U S A. 1996;93:7717–7722. 22. Hofmann C, Obermeier F, Artinger M, et al. Cell–cell contacts prevent anoikis in primary human colonic epithelial cells. Gastroenterology. 2007;132:587–600. 23. Kuhnert F, Davis CR, Wang HT, et al. Essential requirement for Wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expres- sion of Dickkopf-1. Proc Natl Acad Sci U S A. 2004;101:266–271. 24. Ootani A, Li X, Sangiorgi E, et al. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat Med. 2009;15:701–706. 25. Barker N. Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat Rev Mol Cell Biol. 2014;15:19–33. 26. Harkavyi A, Whitton PS. Glucagon-like peptide 1  receptor stimulation as a means of neuroprotection. Br J Pharmacol. 2010;159:495–501. 27. Hayes MR, De Jonghe BC, Kanoski SE. Role of the glucagon-like-peptide-1 receptor in the control of energy balance. Physiol Behav. 2010;100:503–510. 28. Bond A. Exenatide (Byetta) as a novel treatment option for type 2 diabetes mel- litus. Proc (Bayl Univ Med Cent). 2006;19(3):281–284. 29. Wilson CL, Ouellette AJ, Satchell DP, et al. Regulation of intestinal α-defensin activation by the metalloproteinase matrilysin in innate host defense. Science. 1999;286:113–117. 30. Tanabe H, Ayabe T, Bainbridge B, et al. Mouse Paneth cell secretory responses to cell surface glycolipids of virulent and attenuated pathogenic bacteria. Infect Immun. 2005;73:2312–2320. 31. Elphick DA, Mahida YR. Paneth cells: their role in innate immunity and inflammatory disease. Gut. 2005;54(12):1802–1809. 32. Gottesman MM, Pastan I. Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem. 1993;62:385–427. 33. Cole SP, Sparks KE, Fraser K, et al. Pharmacological characterization of multi-drug resistant MRP-transfected human tumor cells. Cancer Res. 1994;54: 5902–5910.