Vol.1, No.3, 44-53 (2011)tem Cell Discovery
doi:10.4236/scd.2011.13005
C
opyright © 2011 SciRes. Openly accessible at http://www.scirp.org/journal/SCD/
S
Endodermal progenitor cells isolat ed from mouse
pancreas
Lisa Samuelson1, Natasha Wright1, David Allen Gerber1,2*
1Department of Surgery, University of North Carolina, Chapel Hill, USA;
2Lineberger Cancer Center, University of North Carolina, Chapel Hill, USA;
*Corresponding Author: david_gerber@med.unc.edu
Received 11 June 2011; revised 23 July 2011; accepted 5 August 2011.
ABSTRACT
In this study we have isolated and identified a
progenitor cell populatio n located in murine pan-
creatic tissue. This cell population has specific
properties that make it different from previously
described candidate pancreatic progenytor cell
populations. It is located in the periductal region
outside the islet architecture yet the cells have
features consistent with endodermal develop-
ment and endocrine functions within the islet.
Cells are proliferative and capable of differentia-
tion into pancreatic lineages expressing pancre-
atic associated transcription factors and proteins
and maintaining the ability to produce insulin.
Keywords: Pancreatic Progenitor; Diabetes; Sca-1;
Murine Pancreas
1. INTRODUCTION
Diabetes has reached epidemic proportions in the U.S.
with an estimated prevalence of 6.3% of the U.S. popu-
lation (translating into more than 18 million people)
currently diagnosed with the disease [1]. Diabetes af-
fected an estimated 171 million people worldwide in
2000, and this number is predicted to rise to 366 million
by 2030, owing to increases in age, obesity, and urbani-
zation of the world’s population [2]. This rising inci-
dence of patients diagnosed with diabetes drives the qu
est for the development of novel therapies.
Diabetes is unique in that there is already a preexist-
ing protocol of transplantation of donor beta cells to di-
abetic recipients which ameliorates the disease. However,
the requirement of large numbers of beta cells and a
shortage of donor tissue limits the procedure to select
cases. The need for a highly proliferative and easily ac-
cessible cell population that can replace transplanted
islets has led to the search for stem cell populations that
can be used to treat the disease. Numerous investigators
have attempted to identify and isolate a pancreatic stem
or progenitor cell using distinct markers [3-7]. While a
great deal of information is known about pancreatic de-
velopment from the embryonic period through adulthood,
there have been tremendous challenges associated with
characterizing a putative pancreatic or islet stem cell.
While the presence of precursor cells has been debated
over the past decade [8-10], the potential presence of a
precursor cell remains a possibility, as demonstrated by
Liu et al via cell tracing experiments [11]. One group has
established a murine pancreatic stem cell line that ex-
presses PDX-1 but does not express select transcription
factors such as Ngn3, BETA2/NeuroD, Pax4, Pax6, Isl-1,
Nkx2.2 or Nkx6.1 [12]. Populations in mouse and hu-
man have been derived from pancreatic ductal cell
components and maintained in long-term culture, where
they could differentiate into multi-lineage cell types
[13-15]. More recent experiments have mechanistically
demonstrated how pancreatic-duct cells can serve as a
source of regeneration [16]. The challenge with many of
these populations was that they were characterized ret-
rospectively after isolation. Petropavlovskaia et al. cha-
racterized a “small cell” isolated from islets that demon-
strates expression of several pancreatic markers (insulin,
glucagon, somatostatin, pancreatic polypeptide, etc.) during
culture but doesn’t express some of the stem cell related
markers (e.g. c-kit, CD34) [17]. Additionally the identi-
fied cells demonstrate a very low level of proliferation.
Other researchers have studied embryonic stem cells
to explore if they could be driven towards a pancreatic
phenotype [18,19]. Some researchers have induced em-
bryonic stem cells to a pancreatic precursor cell based on
the cell’s expression of Insulin I, Insulin II, PDX1 and
other early marker proteins such as peptide YY (PYY)
and glucagon [20,21]. Some investigators have focused
on the expression of Ngn3 as a marker for a pancreatic
progenitor cell during embryogenesis [22]. Select studies
have provided direct evidence that Ngn3+ cells are po-
tential islet progenitors during embryogenesis as well as
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. Openl y accessible at http://www.scirp.org/journal/SCD/
4545
in adult mice, and that Pdx1+ cells give rise to all three
types of pancreatic tissue: exocrine, endocrine and ductal
[22]. Ngn3 is required for specification of pancreatic
endocrine cells along with additional molecules, such as
Notch family members [23,24]. Ngn3+ cells are unipo-
tent at the single-cell level but together as a population
they are multipotent [25]. Other researchers have fo-
cused on fetal pancreatic duct cells as the origin of a
multipotential cell population capable of differentiating
into insulin-producing cells [26]. In addition there have
been studies looking at clonal identification of multipo-
tent precursors from adult mouse pancreas [27,28]. Un-
der further selection, these cells have produced distinct
populations of pancreatic β-, α- and δ-cells, pancreatic
exocrine and stellate cells, and neuronal and glial cells
[27,28]. Additional investigators have looked at non-β-
cell populations (e.g. α-cells, gland cells, etc.) as a source
of potential beta cells [29,30].
We have identified a pancreatic progenitor cell iso-
lated from mouse pancreas. This highly proliferative cell
population is purified based on expression of stem cell
antigen 1 (Sca-1). Cells express many transcription fac-
tors associated with pancreatic development and also
have the ability to produce proteins associated with pan-
creatic endocrine function. These cells also that the abil-
ity to secret a basal amount of insulin.
2. MATERIALS AND METHODS
2.1. Reagents
Chemicals were obtained from the Sigma Chemical
Company (St. Louis, MO) unless otherwise stated. Sca-1
antibody was purchased from BD Biosciences (San Jose,
CA).
2.2. Mice
C57BL/6 mice breeding pairs were purchased from the
Jackson Laboratory (Bar Harbor, ME). All animals were
maintained on a rodent chow under a constant day/night
cycle. 2 - 3 week old mice were used in all experiments.
All care and use of animals was approved by the Institu-
tional Animal Care and Use Committee at the University
of North Carolina at Chapel Hill in accordance with the
principles and procedures outlined in the National Insti-
tutes of Health Guide for the Care and Use of Laboratory
Animals.
2.3. Enrichment, Cell Sorting, and Culture
Pancreatic tissue from 2 week old C57Bl/6 mice was
removed and the tissue was digested with Liberase
Blendzyme 3 (Roche) in a 36˚ water bath for 15 min with
agitation every 5 minutes. The tissue was then sheared
with a 22G needle and tweezers and pipetted until all cells
were disassociated. Cell sorting was performed using
magnetic activated cell sorting (MACS®, Miltenyi, Bio-
tec, Inc.). The cells were separated using a Sca-1 antibo-
dy conjugated to mini-magnetic beads (Miltenyi Biotec
Inc.; Auburn, CA, (http://www.milten yibiotec.com) acc-
ording to the manufacturer’s instructions. Sca1+ cells
were eluted with a purity of >94% by flow cytometry
and >80% viability. Standard culture conditions involved
plating the cells on tissue culture dishes coated with Fi-
bronectin and Concanavalin A in 2 ml of DMEM (Dul-
becco’s Modified Eagle’s medium) and 10% FBS suPple-
mented with 10 ng/ml BMP-4 (R & D Systems), 1400
U/ml ESGRO(Millipore, Temecula, CA), and B27 sup-
plement (Invitrogen, Carlsbad, CA). Cellular density was
1 × 106 cells per 35 mm well. The cells were cultured in a
5% CO2/95% room air incubator at 37˚C. The cells were
passaged when they achieved >75% confluence on the
tissue culture dishes.
2.4. Microscopy
The cells in culture were visualized with either a Zeiss
Axiovert 100 inverted fluorescent microscope and images
were captured with Zeiss Axiovision 3.1 software or an
Olympus BX61 Upright Fluorescence with Improvision’s
Velocity software provided by the Microscopy Services
lab in the department o Molecular and Cellular Pathology.
2.5. Fluorescent Immunophenotyping and
Flow Cytometry
Cells were stained for immunofluorescense using an-
tibodies directly labeled with the relevant fluoroprobe.
Cells were considered positive when fluorescence was
greater than 95% of the negative control cells. Isotype
control antibodies were used as negative controls. Cells
were suspended in room temperature PBS containing 2%
FBS at 1 × 106 cells per ml. Antibodies were primary
conjugates against Sca-1 (BD Biosciences; San Jose, CA).
Cells were incubated for 20 minutes, centrifuged and re-
suspended in cold buffer at 1 × 106 per ml.
2.6. Histology and Immunohistochemical
Characterization
Immunohistochemistry was performed by an indirect
immunoperoxidase procedure for localization of select
proteins (Monga et al., 2001). Primary antibody was a-
gainst Sca-1 (BD Biosciences; San Jose, CA). The secon-
dary antibody was from Vector Laboratories (Burlingame,
CA) and the signal was detected using the ABC Elite and
DAB kits (Vector Laboratories, Burlingame, CA). DAB
staining was toned with DAB Enhancer (Vector Labora-
tories) and counterstained with hemotoxylin. Slides were
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. Openl y accessible at http://www.scirp.org/journal/SCD/
46
blocked with Protein Blocking Agent (Thermo Electron
Solutions; Pittsburgh, PA). For negative control, sections
were incubated with isotype control antibodies. All
stained slides were viewed on the Nikon Microphot-
FXA microscope. It is equipped with an Optronics DEI
750 3-chip CCD camera and a Q Imaging Micropub-
lisher CCD camera for digital image acquisition. Images
were captured on an Apple Power Macintosh G3 com-
puter utilizing Q Imaging software and saved using
Adobe Photoshop CS2 software.
2.7. RT-PCR
Polymerase chain reaction (PCR) analysis was per-
formed on total RNA extracted from isolated Sca-1 posi-
tive and negative cells. A total of 2g of total RNA was
used as a template to create complementary DNA as per
the manufacturer’s instructions for the Retroscript kit
from (Ambion; Austin, TX). The sequences for selected
primers are listed in Supplemental Data Table 1. The
generated complementary DNA was then subjected to a
denaturing temperature of 95˚C for 2 minutes, followed
by further denaturation at 95˚C for 1 min, annealing
temperatures of 55˚C - 70˚C for 45 seconds, and exten-
sion for 60 seconds at 72˚C for 50 cycles, followed by a
final extension at 72˚C for 5 minutes.
Table 1. RT- PCR primers.
Primer 5’-3’ Annealing Temp Size
Ptf1alpha catagagaacgaaccaccctttgag
cttgagacaggtcctttgaggcacg 60 294
Nkx2.2 aaaggtatggaggtgacgcct
tcatgttgcgggtcatgtcga 54 190
Ngn3 aagagcgagttggcactgagc
gcgtatcgcctggtgtcgaa 56 223
Insulin 1 tagtgaccagctataatcagagac
agccaaggtctgaaggtc 70 288
Insulin 2 ccctgctggccctgctctt
aggtctgaaggtcacctgct 60 213
Nestin ggagagtcgcttagaggtgc
gaagagaaccgaaaggactg 58 327
Glut 2 ggataaattcgcctggatga
tcaaggtctttggtttcctt 53 298
NeuroD cttggccaagaactacatctgg
ccgttgaagagaaagtttgtgc 53 430
CK19 ctgcagatgacttcagaacc
ggccatgatctcatactgac 62 299
Pax 6 tcacagcggagtgaatcag
cccaagcaaagatggaag 58 332
Hnf6 gcaatggaagtaattcagggcag
cgtgacagtcgttgaagaagtac 60 471
PDX-1 ctttcccgtggatgaaatcc
gtcaagttcaacatcactgcc 60 205
2.8. Western Blot Analysis
Cells were lysed in buffer containing 50mM Tris,
150mM NaCl, 1% Nonidet P40 (Roche; Indianapolis,
IN), 0.5% deoxycholate, 1mL protease inhibitor cocktail
for every 100 mL (5 μg/mL aprotinin, leupeptin, pep-
statin, and soybean trypsin inhibitor), and 1mL phospha-
tase inhibitor for every 100 mL. Lysates were clarified
by centrifugation at 14,000 g × 2 min and stored at
–20˚C. Protein concentrations were determined by Bio-
Rad protein assay. Total cellular proteins (50 μg/lane)
were dissolved by SDS-PAGE, transferred to nitrocellu-
lose membrane incubated with blocking buffer (5% non-
fat dry milk in 1× Tris Buffered Saline with 0.05%
Tween 20, pH 7.5), and probed with primary antibodies.
After incubation with secondary antibodies, peroxidase
activity was detected by enhanced chemiluminescence.
Densitometric signals from Western blots were analyzed
with NIH-ImageJ software (http://rsb.info.nih.gov/ij/) [31].
Protein levels were calculated in arbitrary units (AU)
normalized with β-actin protein levels.
2.9. Insulin Assa y
Insulin secretion was measured using an enzyme lin-
ked immunosorbent assay (ELISA). Cells were cultured
in either a high or low glucose environment (5 mM and
25 mM respectively) for either a 4 or 48 hour duration.
Supernatant was collected from culture dishes and pla-
ced at –20˚C storage. The samples were allowed only 1
freeze-thaw before analysis. The samples were analyzed
according to the manufacturer’s protocol (Alpco Diagno-
stics, Salem, NH) by the UNC Center for Gastrointestinal
Biology and Disease (CGIBD) Immunotechnology core
facility funded by NIH grant# P30-DK34987and the ins-
ulin concentrations calculated using standard curves ge-
nerated from each assay.
2.10. Cell Proliferation Assay
Cell growth and proliferation were measured using the
CyQUANT® NF Cell Proliferation Assay Kit (Invitrogen
Molecular Probes; Eugene, OR) which measures DNA
content. Cells were allowed to proliferate in 96 well cul-
ture dishes for set time points of 0,1,4,7,14, and 21 days.
On test day growth medium was removed and 50 µl of
1× binding dye from kit added. Cells were further incu-
bated for 30 minutes and then assayed on a BioTek Mi-
croplate reader at emission wavelengths of 485 and 530
nm. Absorbance values obtained were then correlated to
cell numbers using a standard curve. Standard curve was
created previously with a rapidly proliferating cell line.
3. RESULTS
Sca-1 is a marker demonstrated on select stem cell po-
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. http://www.scirp.org/journal/SCD/
4747
pulations including hematopoietic stem cells, endothelial
progenitor cells, adipose stem cells and hepatic stem cells
[32-35]. Pancreatic tissue isolated from newborn and adult
mice, (12 days and 6 weeks old, respectively) was evalu-
ated for the presence of Sca-1+ cells. In newborn mice the
Sca-1+ cells are localized in the periductal region (Figure
1(a)). In adult mice Sca-1+ cells are located in a periductal
and peri-islet distribution (Figure 1(b)) with a noted ab-
sence of Sca-1+ cells within the islet matrix.
Openly accessible at
Pancreatic tissue was subsequently disbursed into a
cellular suspension using a combination of mechanical
and enzymatic separation techniques followed by en-
richment for the Sca-1+ cells (Sca-PPC) with an estab-
lished magnetic separation technique. This isolation te-
chnique generates an average of 1 × 106 Sca-1+ cells from
the pancreas and 5 106 Sca-1 cells. Cellular enrichment
for the Sca-1+ cells was confirmed by flow cytometric
analysis (Figure S1-Supplemental Data).
3.1. Cellular Proliferation and
Characterization
After isolation, the Sca-PPC were placed on tissue cul-
ture dishes using a modification of Ta’s media conditions
[36]. Small colonies of tightly packed cells with large
nuclei are seen on the dishes within the first three days
of culture (Figure 2a). These colonies rapidly expand in
culture forming large colonies. As these cells proliferate
they morphologically change with an increase in cyto-
plasm to nucleus ratio (Figure 2b). Assessing the rate of
cellular proliferation using a mitochondrial assay dem-
onstrates an initial slow proliferation rate of the Sca-PPC
during the first week of culture followed by an accelera-
tion over the subsequent 14 days in culture (Figure 2c).
Due to the periductal location of the Sca-PPC in the pan-
creatic parenchyma we analyzed the tissue for CK19 ex-
pression, a marker associated with ductal cells (e.g. bile
ducts, pancreatic ducts, etc.). (Supplemental Figure S2)
Immunohistochemistry of the pancreatic tissue demon-
strates similar staining patterns for CK19 and Sca-1. We
subsequently evaluated the proliferating Sca-PPC for CK19
expression. Figure 3 demonstrates CK19 expression on a
day 7 colony of proliferating Sca-PPC. During the in vitro
culture period, the Sca-PPC were analyzed for persistence
of Sca-1 expression. Figure 4 is a day 3 colony demon-
strating persistent Sca-expression on the cells. The nuc-
lei of these cells are counterstained with DAPI.
3.2. Transcription Factors and
Differentiation
Transcription factor expression associated with pancr-
eatic development and/or mature pancreatic cells was ev-
aluated in the Sca-PPC population. We selected defined
time points to evaluate cellular differentiation: immedi-
ately after isolation (P0) and in culture after the cells had
been passaged (P2 and P4). Murine islets were used as a
control for these experiments. The Sca-PPC demonstr-
ates expression of Hnf6 at isolation and at later passage,
P4. These cells also expressed Pax6, Nkx2.2, Pdx-1, Ptf1-α,
Ngn3, NeuroD, nestin, CK19, Insulin 1 and 2 but did not
express Glut 2. Figure 5 is a representative set of images
from these experiments. Experiments were performed a
minimum of three times to confirm the presence or ab-
sence of a transcription factor.
Based on the PCR results we assessed Sca-PPC diff-
erentiation by comparing markers associated with the en-
docrine component of the islet and the acinar fraction of
the pancreas. Western blot analyses demonstrate de novo
low expression levels of glucagon and Ngn3 which persist
during cell passage. Insulin receptor-α expression is hig-
her at all three time points in culture, but there are no sta-
tistically significant differences in this experiment (Figure
6). The Sca-PPC also demonstrates de novo expression of
amylase with persistent expression during cell culture.
Pdx-1 is selectively expressed in β-cells and is essential
for maintenance of phenotype in adult β-cells [37]. Based
on the finding of Pdx-1 by PCR (Figure 5) we performed
immunofluorescense analysis of the Sca-PPC colonies to
assess cellular expression. Figure 7 demonstrates strong
Pdx-1 expression on the majority of colony-forming Sca-
PPC.
(b)
(
c
)
(
a
)
Figure 1. Sca-1 expression in normal murine pancreas tissue; (a) Immunohistochemical staining shows Sca-1 expression along the duc-
tal network of neonate mice 2 weeks of age; (b) Expression is also seen along the ductal epithelium as well as in the peri-islet space of
adult mice 6weeks of age; Image (c) represents an isotype control for Sca-1. (a) 20×, (b, c) 10×.
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. Openl y accessible at http://www.scirp.org/journal/SCD/
48
Figure 2. Changes in Sca-1+ colony morphology throughhout
culture. (a) Sca-1+ colony morphology changes over the duration
of standard culture. Initial colonies are similar those seen with
other stem cell sources. Colonies are tightly packed with indi-
vidual small cells each containing a high nuclear to cytoplasm
ratio. Image of day 3 colony. Scale bar 20 µm; (b) By day 21 of
culture colonies spread over 90% of dishes and have a cobble
stone type appearance. Scale bar 50 µm; (c) Proliferation assay to
evaluate doubling time of Sca-PPCs. The proliferation is most
accentuated between days 7 - 21.
Figure 3. (a) CK19 expression of a day 7 colony. Red correlates
with CK19, blue correlates with DAPI staining of nuclei; (b)
Corresponding iso-type control. Both images 10×.
Figure 4. Sca-1 Immunohistochemistry in culture. Colonies in standard culture are formed from Sca-1+ cells, not contaminating cell
types. (a) DAPI staining delineates nuclei within a transmission image of a day 3 colony; (b) Merged DAPI and Sca-1 staining.(c)
Isotype control. (a ,b) 20×, scale bar 20 µm. (c)10×.
3.3. Insulin Production of PPC
Experiments were established to analyze Sca-PPC insulin
production looking at both sustained production and ra-
pid response to environmental changes in glucose con-
centration. The Sca-PPCs demonstrate a basal produ-
ction of insulin using either low or high glucose concen-
trations (P0 cells) while the cells that have been pass-
aged twice (P2) demonstrate an increase in insulin secre-
tion at both low and high glucose concentrations (Figure
8(a)). In the later passage (P4), insulin production is sim-
ilar to the pattern demonstrated when the cells are initi-
ally isolated. These assays are based on a 48 hour col-
lection sample and potentially mask rapid changes in
insulin secretion. We established a rapid response assay
with the Sca-PPC at both low and high glucose concen-
trations and compared them with freshly isolated islets.
Samples were assessed immediately after the media cha-
nge (30 minutes) and several times over a four hour period.
This experiment demonstrates persistent basal insulin pro-
duction by the Sca-PPC without acute responsiveness to
(
a
)
(b)
(
c
)
(
a
)
(b)
(
a
)
(b)
(
c
)
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. Openl y accessible at http://www.scirp.org/journal/SCD/
4949
changes in glucose concentrations within the media (Fi-
gure 8(b)). Sca-PPC insulin production is distinctly dif-
ferent from islets cultured in high glucose concentra-
tion media. Comparatively the Sca-PPC have a higher
production of insulin in low glucose concentration media
experiments compared with islets at the same time points,
30 and 60 minutes respectively (Figure 8b).
4. DISCUSSION
Diabetes is currently the seventh leading cause of
death, the leading cause of kidney failure, lower limb am-
putations, and blindness, as well as a major contributor to
Figure 5. RT-PCR analysis of Sca-1+ PPCs. PCR analysis
shows that PPCs differentiate toward multiple lineages of pan-
creatic fate and not toward simply endocrine β-cell fate. Cells
were assayed at passage 0, 2, and 4 for the listed pancreatic
transcription factors. Ck19 was also analyzed since its expres-
sion was seen in staining in vitro (refer to Figure 3).
Figure 6. Protein expression of Sca-1+ PPCs. Western blot an-
alyses performed at passage numbers 0, 2, 4, and 7 for pancre-
atic proteins Amylase, Insulin Receptor Alpha, Glucagon,
Ngn3, and Pdx-1. Values are reported in arbitrary units. Results
correspond with transcription data in that there is no clear line
of differentiation. No statistical significance was observed.
Error bars reflect standard error. Arbitrary units are normalized
for expression of control protein β-actin.
Figure 7. PDX-1 is expressed by all β-cells of the pancreas and
is necessary in formation of β-cell tissue from progenitor cell
origins. PDX-1 is expressed on day 3 Sca-1+ colonies. Expres-
sion is strongly shown in the cytoplasm and not the nuclear
region of the cells. (a) Transmission image with DAPI deline-
ating nuclei. (b) PDX-1 expression of the same colonies using
PE conjugated antibody. (c) Isotype control. All images 10×.
Scale bar for (a) and (B) equals 20 µm.
(
a
)
(b)
(c)
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. Openl y accessible at http://www.scirp.org/journal/SCD/
50
Figure 8. Functional ability of Sca-1+ PPCs to produce insulin.
(Top panel) ELISA was utilized to measure sustained secretion of
insulin by neonate (2 week) Sca-1+ donor cells. Cells were stimu-
lated with low (2.8 mM) and high glucose (40 mM) conditions for
a 48 hour period and supernatant collected at the conclusion. (Bot-
tom panel) Insulin ELISA was also utilized to measure rapid re-
sponse to increased glucose conditions. Neonatal cells were stim-
ulated for a 4 hour duration with either high (40 mM) or low (5.55
mM) glucose conditions in the absence of FBS. Supernatants were
collected at 30 min, 1, 2, and 4 hours. Cells were assayed at Pass-
age 0 of culture during the time period when small colonies are
visible. Mature islets were used as controls. Units are expressed in
ng/ml normalized for 50 µg of protein. No statistical significance
was observed. No statistical significance was observed. Error bars
represent standard error.
heart disease and stroke in the US [38]. Current treatment
options for Type 1 diabetics do not offer physiologi-
cally-responsive strict control of blood sugar levels to
prevent the development of the previously mentioned
complications. Islet cell transplant is a clinically proven
method to treat Type I diabetes but it is challenged by
severe limitations in donor availability, poor long term
success rate, and the requirement for immunosuppres-
sion. For these reasons an alternative cell source that
could provide glucose responsiveness offers a potential
cure to the approximately 2 million newly diagnosed T1D
patients each year in the US and the T2D insulin depend-
ent patients [38].
Ongoing debate in the field of stem cell biology ques-
tions the existence of a progenitor cell in adult pancrea-
tic tissue. Initial studies have shown that new β-cells
arise as a result of division of pre-existing β-cells throu-
ghout life and after pancreas injury, thus casting doubt
on the idea of stem cell contributions [9,10,39,40]. This
contrasts with others who have shown that a stem cell
does exist in the pancreas. Seaberg et al. isolated pan-
creatic precursors from adult mouse pancreas and dem-
onstrated that the cells are positive for many β-cell
markers and capable of releasing insulin in response to
glucose stimulation [28]. However, the amount of insulin
produced by these cells was much lower than what is
produced by a naturally occurring islet-derived β-cell. It
has also been shown that endocrine precursors can be
derived from surgically resected portions of human pan-
creas and that islets themselves contain a population of
mesenchymal stem cells [41,42]. Recently published
data bolsters the idea of a precursor cell in pancreas cit-
ing the existence of insulin positive multipotent stem
cells that give rise to pancreatic and neural lineages and
that ameliorate diabetes in animal models [43].
Novel cell surface antibodies are being developed that
allow the selective isolation of individual cell popula-
tions, (e.g. exocrine cells) providing a useful tool to stu-
dy adult derived progenitors [44]. Our current study de-
monstrates the presence of a tissue derived progenitor
cell from murine pancreas that is highly proliferative and
able to be purified using a select cell surface marker,
Sca-1+. Other reports demonstrate Sca-1 expression in
progenitor cells of the kidney, liver, cardiac and skeletal
muscle, and mammary tissue but to our knowledge this
is the first report of a Sca-1+ progenitor cell residing in
murine pancreas [34,45-48].
CK19 is another marker found on the Sca-PPCs dur-
ing in vivo and in vitro assessment. CK19 is a ductal
marker and its expression in murine pancreas is found to
overlap with ductal areas positive for Sca-1. In early cell
culture experiments all of the Sca-PPC colonies are posi-
tive for CK19. This expression of CK19 is important in
translating our research to other species. Sca-1 is an an-
tigen that is specific to murine tissues thus an alternative
marker is needed when we analyze human tissue. Sev-
eral groups have reported the possibility of multipotent
stem cells associated with ductal areas and the biliary
ducts that traverse the pancreas, [16,49,50]. The pres-
ence of CK19 in our Sca-PPCs may indicate overlap
with other isolated precursor cells and offer an opportu-
nity for exploration of new antigens of interest.
Analysis of cultured Sca-PPCs demonstrates variabil-
ity in the expression of differentiation markers. The cells
initially express many of the developmental markers ass-
ociated with pancreatic and endocrine progenitor cells, in-
cluding Hnf-6, Ptf1α, PDX-1, Pax6, Nkx2.2, Ngn3, Neu-
roD, and Nestin. Variability in transcription factor expres-
sion is demonstrated throughout the periods of culture
that were studied. After initially expressing multiple en-
docrine developmental markers the Sca-PPCs lose expr-
ession of Hnf6 and Pax6 while maintaining expression
of Nkx2.2, Ngn3 and Neuro D. Cells also express differ-
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. Openl y accessible at http://www.scirp.org/journal/SCD/
5151
entiated cell markers including Insulin1, Insulin2, Glut2,
and CK19. These markers are initially present but by the
time the cells have undergone multiple passages the cells
lose expression of both Insulin 1 and 2. A possible expl-
anation for the variability of expression could relate to
cell-cell interactions in the in vitro cellular environment
and the impact on the cells, as cells cultured on a rigid
surface have been previously associated with cellular di-
fferentiation [51,52]. Maintaining the cells in a 2-D env-
ironment thereby limits their ability to develop normally
and forces the cells to differentiate based on constraints
of the culture vessel as well as causing cell death due to
limited space.
PDX-1 is a transcription factor that is present during
early development of the pancreas and is essential for the
differentiation process into mature β-cells. For this reason
its presence would theoretically be essential in any cell
population which has the potential of becoming a func-
tional islet-like cell. Sca-PPCs express PDX-1 throughout
culture at the transcriptional level although in vitro analy-
sis demonstrates its expression in the cells’ cytoplasm.
This is distinct from the typical nuclear location of PDX-
1. This cytoplasmic localization in the PPCs suggests sig-
nal transcription but not translation of the protein into an
active form. This observation has been demonstrated by
other investigators where PDX-1 is found in an inactive
form in the cytoplasm of the islet when glucose concen-
trations are low. PDX-1 is then activated by phosphory-
lation when glucose concentrations increase and the ac-
tive form is translocated to the cell nucleus [53-56].
Protein analysis of the Sca-PPC population shows that
cells express the developmental markers Ngn3 and PDX-1
throughout the analyzed periods of culture. The cells
also express mature endocrine proteins glucagon and In-
sulin Receptor α. Likewise, the mature exocrine cell ma-
rker amylase is expressed throughout the in vitro culture
periods. The expression levels of each protein vary thr-
oughout the experiment showing the cells’ ability for en-
docrine and exocrine functions.
Sca-PPCs have the functional ability to produce large in
vitro amounts of insulin. This is a distinct finding com-
pared to published studies where insulin secretion is very
limited in progenitor cell populations. Sca-PPCs from
adult mice produce a steady state of insulin across passa-
ges and over a 2 day period of culture yet the release is
independent of changes in glucose concentration. Neona-
tal mice are capable of responding to glucose concentra-
tion changes at passage 2, but do not maintain this ability
in extended culture. In acute phase experiments there is
no increase in insulin secretion from the Sca-PPCs in
high glucose conditions over a four hour period. In fact,
cells exposed to low glucose conditions released more
insulin than their high glucose counterparts. This was in
stark contrast to islets which show a drastic increase in
insulin secretion at high concentrations and only a minor
increase in low glucose conditions. Finding a way to di-
rect the secretion of insulin in response to glucose stimu-
lation in the Sca-PPC’s may show that they have potential
as a stem cell source capable of treating diabetes.
This study demonstrates the presence of a Sca-1+ pro-
genitor cell subset in the murine pancreas. The expres-
sion of CK19 offers a potential complementary marker
for selection of a correlating population in human pancr-
eatic tissue. This co-expression of ductal markers may
also offer insight to the origin of these progenitor cells.
Based upon the common embryologic development of
pancreas and liver from the foregut endoderm the compl-
ex ductal networks of both organs likely share a com-
mon origin. This is corroborated by the fact that our lab
previously identified a Sca-1+ cell residing in adult mu-
rine liver [34]. Regulation of the proliferative and diff-
erentiation features of this cell population will be critical
as we continue to investigate alternative strategies for
glucose regulation in our diabetic population.
REFERENCES
[1] Engelgau, M.M., et al. (2004) The evolving diabetes
burden in the United States. Annals of Internal Medicine,
140, 945-950.
[2] Diabetes Prevention Program Research Group (2009) 10-
year follow-up of diabetes incidence and weight loss in
the Diabetes Prevention Program Outcomes Study. The
Lancet, 374, 1677-1686.
doi:10.1016/S0140-6736(09)61457-4
[3] Bonner-Weir, S. and Sharma, A. (2002) Pancreatic stem
cells. Journal of Pathology, 197, 519-526.
doi:10.1002/path.1158
[4] Teitelman, G., et al. (1993) Precursor cells of mouse en-
docrine pancreas coexpress insulin, glucagon and the
neuronal proteins tyrosine hydroxylase and neuropeptide
Y, but not pancreatic polypeptide. Development, 118,
1031-1039.
[5] Zulewski, H., et al. (2001) Multipotential nestin-positive
stem cells isolated from adult pancreatic islets differenti-
ate ex vivo into pancreatic endocrine, exocrine, and he-
patic phenotypes. Diabetes, 20, 521-533.
doi:10.2337/diabetes.50.3.521
[6] Schwitzgebel, V.M., et al. (2000) Expression of neuro-
genin3 reveals an islet cell precursor population in the
pancreas. Development, 127, 3533-3542.
[7] Rovira, M., et al. (2009) Isolation and characterization of
centroacinar/terminal ductal progenitor cells in adult
mouse pancreas. Proceedings of the National Academy of
Sciences of the USA, 107, 75-80.
doi:10.1073/pnas.0912589107
[8] Dor, Y. and Melton, D.A. (2008) Facultative endocrine
progenitor cells in the adult pancreas. Cell, 132, 183-184.
doi:10.1016/j.cell.2008.01.004
[9] Brennand, K., Huangfu, D. and Melton, D. (2007) All
beta cells contribute equally to islet growth and mainte-
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. Openl y accessible at http://www.scirp.org/journal/SCD/
52
nance. PLoS Biology, 5, e163.
doi:10.1371/journal.pbio.0050163
[10] Dor, Y., et al. (2004) Adult pancreatic beta-cells are
formed by self-duplication rather than stem-cell differen-
tiation. Nature, 429, 41-46. doi:10.1038/nature02520
[11] Liu, H., et al. (2010) Precursor cells in mouse islets gen-
erate new {beta}-cells in vivo during aging and after islet
injury. Endocrinology, 151, 520-528.
doi:10.1210/en.2009-0992
[12] Noguchi, H., et al. (2009) Establishment of mouse pan-
creatic stem cell line. Cell Transplant, 18, 563-571.
[13] Cornelius, J.G., et al. (1997) In vitro generation of islets
in long-term cultures of pluripotent stem cells from adult
mouse pancreas. Hormone & Metabolic Research, 29,
271-277. doi:10.1055/s-2007-979036
[14] Ramiya, V.K., et al. (2000) Reversal of insulin-dependent
diabetes using islets generated in vitro from pancreatic
stem cells. Nature Medicine, 6, 278-282.
doi:10.1038/73128
[15] Bonner-Weir, S., et al. (2000) In vitro cultivation of hu-
man islets from expanded ductal tissue. Proceedings of
the National Academy of Sciences of the USA, 97, 7999-
8004. doi:10.1073/pnas.97.14.7999
[16] Li, W.C., et al. (2010) Activation of pancreaticduct de-
rived progenitor cells during pancreas regeneration in
adult rats. Journal of Cell Science, 123, 2792-2802.
doi:10.1242/jcs.065268
[17] Petropavlovskaia, M. and Rosenberg L. (2002) Identifi-
cation and characterization of small cells in the adult
pancreas: Potential progenitor cells? Cell and Tissue Re-
search, 310, 51-58. doi:10.1007/s00441-002-0614-z
[18] Kroon, E., et al. (2008) Pancreatic endoderm derived
from human embryonic stem cells generates glucosere-
sponsive insulin-secreting cells in vivo. National Biotech-
nology, 26, 443-452. doi:10.1038/nbt1393
[19] D’Amour, K.A., et al. (2006) Production of pancreatic
hormone-expressing endocrine cells from human em-
bryonic stem cells. National Biotechnology, 24, 1392-
1401.
[20] Kahan, B.W., et al. (2003) Pancreatic precursors and
differentiated islet cell types from murine embryonic
stem cells: An in vitro model to study islet differentiation.
Diabetes, 52, 2016-2024. doi:10.2337/diabetes.52.8.2016
[21] Ku, H.T., et al. (2004) Committing embryonic stem cells
to early endocrine pancreas in vitro. Stem Cells, 22,
1205-1217. doi:10.1634/stemcells.2004-0027
[22] Gu, G., Dubauskaite, J. and Melton, D.A. (2002) Direct
evidence for the pancreatic lineage: NGN3+ cells are is-
let progenitors and are distinct from duct progenitors.
Development, 129, 2447-2457.
[23] Apelqvist, A., et al. (1999) Notch signalling controls
pancreatic cell differentiation. Nature, 400, 877-881.
doi:10.1038/21913
[24] Jensen, J., et al. (2000) Control of endodermal endocrine
development by Hes-1. Nature Genetics, 24, 36-44.
doi:10.1038/71657
[25] Desgraz, R. and Herrera, P.L. (2009) Pancreatic neuro-
genin 3-expressing cells are unipotent islet precursors.
Development, 136, 3567-3574. doi:10.1242/dev.039214
[26] Yao, Z.X., et al. (2004) In vitro cultivation of human
fetal pancreatic ductal stem cells and their differentiation
into insulin-producing cells. World Journal of Gastroen-
terology, 10, 1452-1456.
[27] Suzuki, A., Nakauchi, H. and Taniguchi, H. (2004) Pro-
spective isolation of multipotent pancreatic progenitors
using flow-cytometric cell sorting. Diabetes, 53, 2143-
2152. doi:10.2337/diabetes.53.8.2143
[28] Seaberg, R.M., et al. (2004) Clonal identification of mul-
tipotent precursors from adult mouse pancreas that gen-
erate neural and pancreatic lineages. Nature Biotechnol-
ogy, 22, 1115-1124. doi:10.1038/nbt1004
[29] Thorel, F., et al. (2010) Conversion of adult pancreatic
alpha-cells to beta-cells after extreme beta-cell loss. Na-
ture, 464, 1149-1154. doi:10.1038/nature08894
[30] Strobel, O., et al. (2010) Pancreatic duct glands are dis-
tinct ductal compartments that react to chronic injury and
mediate shh-Induced metaplasia. Gastroenterology, 138,
1166-1177. doi:10.1053/j.gastro.2009.12.005
[31] Abramoff, M.D., Magelhaes, P.J. and Ram, S.J. (2004)
Image processing with image. Journal of Biophotonics
International, 11, 36-42.
[32] Khoo, C.P., et al. (2009) Characterization of endothelial
progenitor cells in the NOD mouse as a source for cell
therapies. Diabetes/Metabolism Research and Reviews,
25, 89-93. doi:10.1002/dmrr.898
[33] Chandra, V., et al. (2009) Generation of pancreatic hor-
mone-expressing islet-like cell aggregates from murine
adipose tissue-derived stem cells. Stem Cells, 27, 1941-
1953. doi:10.1002/stem.117
[34] Wright, N., et al. (2008) Enrichment of a bipotent hepatic
progenitor cell from naive adult liver tissue. Biochemical
and Biophysical Research Communications, 366, 367-
372. doi:10.1016/j.bbrc.2007.11.129
[35] Holmes, C. and Stanford, W.L. (2007) Stem cell anti-
gen-1: Expression, function, and enigma. Stem Cells, 25,
1339-1347. doi:10.1634/stemcells.2006-0644
[36] Ta, M., et al. (2006) The defined combination of growth
factors controls generation of long-term-replicating islet
progenitor-like cells from cultures of adult mouse pan-
creas. Stem Cells, 24, 1738-1749.
doi:10.1634/stemcells.2005-0367
[37] Gu, G., Brown, J.R. and Melton, D.A. (2003) Direct li-
neage tracing reveals the ontogeny of pancreatic cell
fates during mouse embryogenesis. Mechanisms of De-
velopment, 120, 35-43.
doi:10.1016/S0925-4773(02)00330-1
[38] Centers for Disease Control and Prevention, (2011) Na-
tional diabetes fact sheet: National estimates and general
information on diabetes and prediabetes in the United
States. Atlanta, GA.
[39] Nir, T., Melton, D.A. and Dor, Y. (2007) Recovery from
diabetes in mice by beta cell regeneration. Journal of
Clinical Investigation, 117, 2553-2561.
doi:10.1172/JCI32959
[40] Teta, M., et al. (2007) Growth and regeneration of adult
beta cells does not involve specialized progenitors. De-
velopmental Cell, 12, 817-826.
doi:10.1016/j.devcel.2007.04.011
[41] Shyu, J.F., et al. (2011) Alleviation of hyperglycemia in
diabetic rats by intraportal injection of insulin-producing
cells generated from surgically resected human pancre-
atic tissue. Journal Endocrinol, 208, 233-244.
[42] Carlotti, F., et al. (2011) Isolated human islets contain a
distinct population of mesenchymal stem cells. Islets, 2,
L. Samuelson et al. / Stem Cell Discovery 1 (2011) 44-53
Copyright © 2011 SciRes. http://www.scirp.org/journal/SCD/
5353
164-173.
[43] Smukler, S.R., et al. (2011) The adult mouse and human
pancreas contain rare multipotent stem cells that express
insulin. Stem Cell, 8, 281-293.
doi:10.1016/j.stem.2011.01.015
Openly accessible at
[44] Dorrell, C., et al. (2011) Isolation of mouse pancreatic
alpha, beta, duct and acinar populations with cell surface
markers. Molecular and Cellular Endocrinology, 399,
144-150.
[45] Dekel, B., et al. (2006) Isolation and characterization of
nontubular sca-1+lin-multipotent stem/progenitor cells
from adult mouse kidney. Journal of the American Soci-
ety of Nephrology, 17, 3300-3314.
doi:10.1681/ASN.2005020195
[46] Oh, H., et al. (2003) Cardiac progenitor cells from adult
myocardium: Homing, differentiation, and fusion after
infarction. Proceedings of the National Academy of Sci-
ences of the USA, 100, 12313-12318.
doi:10.1073/pnas.2132126100
[47] Asakura, A., et al. (2002) Myogenic specification of side
population cells in skeletal muscle. Journal of Cell Biol-
ogy, 159, 123-134. doi:10.1083/jcb.200202092
[48] Welm, B.E., et al. (2002) Sca-1 (pos) cells in the mouse
mammary gland represent an enriched progenitor cell
population. Developmental Biology, 245, 42-56.
doi:10.1006/dbio.2002.0625
[49] Cardinale, V., et al. (2010) Multipotent stem cells in the
biliary tree. Italian Journal of Anatomy and Embryology,
115, 85-90.
[50] Nagaya, M., et al. (2009) Adult mouse intrahepatic bil-
iary epithelial cells induced in vitro to become insu-
lin-producing cells. Journal of Endocrinology, 201, 37-
47. doi:10.1677/JOE-08-0482
[51] Darling, E.M. and Athanasiou, K.A. (2005) Retaining
zonal chondrocyte phenotype by means of novel growth
environments. Tissue Engineering, 11, 395-403.
doi:10.1089/ten.2005.11.395
[52] Ayala, P., Lopez, J.I. and Desai, T.A. (2010) Micro-
topographical cues in 3D attenuate fibrotic phenotype
and extracellular matrix deposition: Implications for tis-
sue regeneration. Tissue Engeneering Part A, 16, 2519-
2527. doi:10.1089/ten.tea.2009.0815
[53] Macfarlane, W.M., et al. (1999) Glucose stimulates trans-
location of the homeodomain transcription factor PDX1
from the cytoplasm to the nucleus in pancreatic beta-cells.
Journal of Biological Chemistry, 274, 1011-1016.
doi:10.1074/jbc.274.2.1011
[54] Andrali, S.S., et al. (2008) Glucose regulation of insulin
gene expression in pancreatic beta-cells. Biochemical
Journal, 415, 1-10. doi:10.1042/BJ20081029
[55] Guillemain, G., et al. (2004) Importin beta1 mediates the
glucose-stimulated nuclear import of pancreatic and du-
odenal homeobox-1 in pancreatic islet beta-cells (MIN6).
Biochemical Journal, 378, 219-227.
doi:10.1042/BJ20031549
[56] Rafiq, I., et al. (2000) Glucose-stimulated preproinsulin
gene expression and nuclear trans-location of pancreatic
duodenum homeobox-1 require activation of phosphati-
dylinositol 3-kinase but not p38 MAPK/SAPK2. Journal
of Biological Chemistry, 275, 15977-15984.
doi:10.1074/jbc.275.21.15977
Supplemental Images
Figure S1. Flow cytometry analysis of immuno-magnetically
separated Sca-1+ PPCs. MACS separation generates a reliable
Sca-1+ cell population. Flow cytometry analysis shows unla-
beled cells (blue diagonal hash lines) as a distinct population
from PE labeled Sca+ cells (red vertical hashed lines), verifying
purity in magnetic separation.
(b)
(a)
Figure S2. (a) Cytokeratin19 is a common marker of ductal
epithelium and serves as a control to delineate the rich ductal
network of the pancreas; (b) Corresponding isotype control.
Both images 20×.