Sie sind auf Seite 1von 16

Biofabrication

PAPER

Sprouting angiogenesis in engineered pseudo islets


To cite this article: Monika Hospodiuk et al 2018 Biofabrication 10 035003

View the article online for updates and enhancements.

This content was downloaded from IP address 193.255.135.254 on 02/10/2018 at 19:13


Biofabrication 10 (2018) 035003 https://doi.org/10.1088/1758-5090/aab002

PAPER

Sprouting angiogenesis in engineered pseudo islets


RECEIVED
7 August 2017
REVISED
Monika Hospodiuk1,2, Madhuri Dey1,3, Bugra Ayan1,4, Donna Sosnoski1,4, Kazim K Moncal1,4,
27 January 2018 Yang Wu1,4 and Ibrahim T Ozbolat1,4,5,6
1
ACCEPTED FOR PUBLICATION The Huck Institutes of the Life Sciences, Penn State University, State College, PA 16801, United States of America
16 February 2018 2
Department of Agriculture and Biological Engineering, Penn State University, State College, PA 16801, United States of America
3
PUBLISHED Department of Chemistry, Penn State University, State College, PA 16801, United States of America
16 March 2018 4
Engineering Science and Mechanics Department, Penn State University, State College, PA 16801, United States of America
5
Biomedical Engineering Department, Penn State University, State College, PA 16801, United States of America
6
Materials Research Institute, Penn State University, State College, PA 16801, United States of America
E-mail: ito1@psu.edu

Keywords: islets, type-1 diabetes, angiogenesis, sprouting, pancreas


Supplementary material for this article is available online

Abstract
Despite the recent achievements in cell-based therapies for curing type-1 diabetes (T1D), capillariza-
tion in beta (β)-cell clusters is still a major roadblock as it is essential for long-term viability and
function of β-cells in vivo. In this research, we report sprouting angiogenesis in engineered pseudo
islets (EPIs) made of mouse insulinoma βTC3 cells and rat heart microvascular endothelial cells
(RHMVECs). Upon culturing in three-dimensional (3D) constructs under angiogenic conditions,
EPIs sprouted extensive capillaries into the surrounding matrix. Ultra-morphological analysis
through histological sections also revealed presence of capillarization within EPIs. EPIs cultured in 3D
constructs maintained their viability and functionality over time while non-vascularized EPIs, without
the presence of RHMVECs, could not retain their viability nor functionality. Here we demonstrate
angiogenesis in engineered islets, where patient specific stem cell-derived human beta cells can be
combined with microvascular endothelial cells in the near future for long-term graft survival in T1D
patients.

1. Introduction derived ECs within the transplant. Revascularization


within the first few days is critical for islets to regain
Type-1 diabetes (T1D) is a devastating disease caused function [7].
by malfunction or complete loss of insulin production Cells in islets are arranged in clusters of 100–1000
by beta (β)-cells in islets of Langerhans in pancreas cells representing less than 1% of the pancreatic mass.
[1, 2]. As a result, insulin is produced minimally or not They receive as much as 15%–20% of total blood flow
at all. In most cases, T1D is caused by an autoimmune to pancreas to regulate homeostasis in the organism
response, whereby the immune system attacks β-cells [10, 11]. β-cells are the only cell type in the organism
and destroys them [1]. It is a chronic disease that often capable of insulin production; therefore, they must
leads to severe complications including blindness, maintain a delicate balance between function and self-
limb amputations, kidney failure, neuropathy, and regeneration. Evidence suggests that ECs directly
cardiovascular diseases [3–5]. Until now, T1D has impact β-cell survival as well as insulin gene expres-
been managed by subcutaneous insulin injections and sion [12]. Insulin secreted by β-cells up-regulates
cure has been attempted by the transplantation of synthesis of endothelial nitric oxide, which promotes
cadaveric pancreases or islets [6]. The crucial aspect of intra-islet blood flow [12, 13]. Also, β-cells secrete
the successful islet transplantation is vascularization endothelial growth factor-A (VEGF-A), which is cru-
[7, 8]. The islets are not directly connected to the blood cial for β-cells development, intra-islet vasculariza-
supply, contrary to transplanted solid organs [9]. tion, and maintenance of β-cell mass [14]. Moreover,
Revascularization depends upon local recruitment of VEGF-A induces proliferation and migration of ECs
endothelial cells (ECs) of the recipient and donor islet- in the developing pancreas, and is strictly controlled to

© 2018 IOP Publishing Ltd


Biofabrication 10 (2018) 035003 M Hospodiuk et al

maintain appropriate intra-islet vasculature and func- Aldrich, St. Louis, MO), 1 μg ml−1 human epidermal
tional architecture [14]. The extracellular matrix growth factor (Sigma-Aldrich), 12 μg ml−1 bovine
(ECM) of islets includes laminin, collagen type IV, and brain extract (Lonza, Walkersville, MD), 100 U ml−1
perlecan, and serves as a physical barrier for infiltra- penicillin G, and 100 μg ml−1 streptomycin. Cells
tion of immune cells. The intra-islet ECs contribute to were maintained at 37 °C in a 5% CO2 humidified
the ECM directly by synthesizing the basement mem- atmosphere. Cell culture medium was changed every
brane [15]. 2–3 d. Subconfluent cultures were detached from the
The current research has focused on engineering flasks using a 0.25% trypsin-0.1% EDTA solution (Life
pancreatic β-cells with ability to secrete insulin Technologies) and split to maintain cell growth.
[16, 17]. The monolayer co-culture of human embryo- Passages 9 through 15 and 10 through 22 were used for
nic stem cells and ECs resulted in specific differentia- βTC3 cells and RHMVECs, respectively.
tion to pancreatic β-cells, even without additional
chemical factors [18]. It allowed for recapitulating 2.2. Mold fabrication for 3D culture
interactions between cells in the developmental niche Negative patterns were designed with cylindrical
that is closely aligned with islet organogenesis. Also, a micro-pillars of different diameters on top of the mold
study by Johannson et al [19] on rat islets indicated surface. PTC Creo software (Parametric Technology
that ECs could affect β-cell function and in vitro stu- Corporation, Exton, PA) was used to create a 3D
dies showed that human umbilical vein ECs co-cul- computer aided design (CAD) model. The CAD model
tured with rat insulinoma cells (INS-1) enhanced was then converted to a stereolithography file to
insulin secretion compared to the INS-1-alone culture fabricate the mold using a Perfactory® Micro Hi-Res
[20]. Also, rotational co-culture of human β-cells and 3D printer (EnvisionTec, Detroit, MI). A high-resolu-
EC demonstrated increased insulin secretion [21]. tion material, HTM140M, (EnvisionTec) was used to
Researchers have already shown pancreatic pseudo 3D print the mold (figure 1(A)). The cylindrical
islets using induced pluripotent stem cells [17], β-cells micro-pillars were 300 μm in diameter with a total
from rodent islets co-cultured with endothelial pro- number of 124 wells. After 3D printing the HTM140M
genitor cells [22], and β-cells from rodent cell line co- negative mold, it was detached from the base, washed
cultured with islet-derived ECs [23]. To our best with detergent, then with 70% ethanol, and sterilized
knowledge, no study has demonstrated the angiogenic under ultraviolet light for 15 min. To cast the mold,
potential of pseudo islets and impact of capillarization 1.5% (w/v) agarose (RPI Corp., Mt. Prospect, IL) was
on islet viability and function. dissolved in MilliQ water, then autoclaved. Liquefied
We here report the sprouting angiogenesis in β- agarose was poured carefully into the negative mold
cell clusters in a three-dimensional (3D) hydrogel cul- and left to solidify for 1 h at room temperature; agarose
ture. The murine pancreatic beta cell line (β-TC3 cells) molds were then detached from the wall of 3D printed
and rat heart microvessel endothelial cells form with needle, and removed by strong concussion
(RHMVECs) were grown in clusters similar to the islet into 100 mm Petri dishes. Prepared agarose molds
structure recreating a native-like environment. These were incubated for 10 min in two changes of appro-
clusters, henceforth referred as engineered pseudo priate culture medium and then stored under sterile
islets (EPIs), sprouted capillaries into the hydrogel as conditions at 4 °C.
well as facilitated neovascularization, which, in turn,
increased the viability and function of β-cell clusters
2.3. Fabrication of EPIs
over time.
On reaching 70% confluence, βTC3 cells and
RHMVECs were detached from cell culture flasks
2. Materials and methods using trypsin; cell media was added to deactivate
trypsin and suspension was centrifuged for 5 min at
2.1. Cell culture 1,600 rpm. Cells were counted using a hemocyt-
Mouse insulinoma beta (β) TC3 cells (βTC3s), a kind ometer. βTC3 and RHMVECs were combined in
gift from Dr Nicolas Zavazava’s laboratory (The ratios of 2:1 or 1:1, respectively. Also a third ratio was
University of Iowa, Iowa City, IA), were cultured in created by βTC3 cells only as a control group. A total
Dulbecco’s Modified Eagle’s Medium (DMEM; Corn- of 2×106 cells were suspended in 100 μl of medium
ing Cellgro, Manassas, VA) supplemented with 20% and carefully pipetted on the top chamber of the
fetal bovine serum (Life Technologies, Grand Island, agarose mold. Gravity acted to pull cells down into the
NY), 1 mM sodium pyruvate (Life Technologies), agarose wells and cells then aggregated as explained
2 mM Glutamax (Life Technologies), and 100 U ml−1 previously [24] (figure 1(B)). Over the next 9 h, at 3 h
penicillin G, 100 μg ml−1 streptomycin (Life Technol- intervals, a small amount (∼150 μl) of fresh medium
ogies). RHMVECs (VEC Technologies, Rensselaer, was gently added to the top of the mold to provide
NY) were cultured in MCDB 131 medium (Corning nutrition for the developing EPIs. The Petri dish
Cellgro) supplemented with 5% fetal bovine serum, containing the molds was also filled with cell culture
2 mM Glutamax, 1 μg ml−1 hydrocortisone (Sigma- media to ensure proper hydration of agarose molds.

2
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Figure 1. EPI fabrication and morphology. (A) 3D printed negative mold for agarose casting. (B) Agarose mold in a Petri dish
surrounded by the cell culture media (top), an EPI on the top view (bottom, left), the cutaway view of an agarose mold showing EPI at
the bottom of a micro-well on Day 3 (bottom, right). (C) Surface tension of βTC3-only, 2:1, and 1:1 groups EPIs measured at Day 1, 5,
and 10 (n=3).

After 12 h, EPIs compacted and media was changed Surface tension of the βTC3-only, 2:1, and 1:1 groups
every 24 h. were measured at Days 1, 5, and 10.

2.4. Morphological analysis 2.6. Scanning electron microscopy (SEM) imaging


The βTC3-only, 2:1, and 1:1 group EPIs were assayed Field emission SEM (Zeiss SIGMA VP-FESEM) was
for size and proliferation at three time points: 1, 5, and used to investigate the surface topography of EPIs.
10 d. At each time point, the size of 15 random EPIs for EPIs were harvested after 3 d of culture in the agarose
three cell ratios in concentrations of 1, 2, and 3×106 mold and fixed in 4% paraformaldehyde (Sigma-
cells/mold was measured using an EVOS FL Auto Aldrich) overnight. EPIs were then carefully washed in
inverted microscope (ThermoFisher, Pittsburgh, PA) PBS and dehydrated using graded ethanol solutions
and software in bright field mode. The relative (25%–100%). To ensure complete removal of water,
difference in average diameter between the 1st (V1) EPIs were then further dried in a critical point dryer
and 10th (V2) day was determined by the equation: (CPD300, Leica EM, Wetzlar, Germany). On complete
(V2 - V1)
∣ V1 ∣
´ 100. The morphology of EPIs was deter- dehydration, EPIs were sputter coated with gold using
mined by razor cutting of the agarose mold containing the Bal-tec SCD-050 Sputter Coater (Leica) and
EPIs. EPIs were observed and imaged on the EVOS FL observed at an accelerating voltage of 3 kV.
Auto inverted light microscope to visualize mold
characteristics and EPI morphology vertically 2.7. Flow cytometry
dimension. In this study, flow cytometry was performed in order
to quantify the ratio of βTC3 cells to RHMVECs of
2.5. Surface tension measurement cultured free-standing EPIs. Prior to analysis,
In order to measure the surface tension of EPIs, a RHMVEC were transduced with EF1 tdTomato lenti-
micropipette aspiration technique was performed vector (Vectalys, Toulouse, France) by 5 h incubation
according to a previous study [25]. The customized at 37 °C in a 6-well plate. The fluorescence-positive
straight micropipette was fabricated from a borosili- cells were sorted using a MoFlo Astrios sorter
cate glass Pasteur pipet (VWR, 14673-043, Radnor, (Brckman Coulter, Pasadena, CA). The brightest cells
PA) using a P97 Flaming/Brown micropipette puller were gated (52.1%, shown in supplementary figure 1 is
(Sutter Instrument, Novato, CA). Aspirated EPIs were available online at stacks.iop.org/BF/10/035003/
monitored via STC-MC33USB monochromatic cam- mmedia) and collected for further cell culture. EPIs in
era (Sentech, Japan) equipped with 1-61448 and 2:1 and 1:1 ratios after 1-, 5-, and 10-day culture were
1-61449 adaptor tubes (Navitar, Rochester, NY). collected in 1.5 ml tubes and trypsinized for

3
Biofabrication 10 (2018) 035003 M Hospodiuk et al

approximately 30 min until segregated to single-cell maximum viability for both cells (data not shown
suspension. Cells were then collected through centri- here). Fabricated fibrin constructs were installed into a
fugation at 1500 rpm for 5 min and fixed overnight live cell imaging chamber and observed using a
with 4% formaldehyde solution (Sigma-Aldrich). Keyence BZ-9000E microscope (Keyence Corp, Bos-
Suspension of cells at each ratio and time point were ton, MA). Images were captured every hour for a 64 h
stained with guinea pig anti-insulin antibody (Ab7842, period.
Abcam) followed by the goat anti-guinea pig second-
ary antibody (Alexa Fluor 488, Life Technologies) to
label βTC3 cells. Sample analysis was carried out using 2.10. Cell viability and proliferation
a LSR Fortessa Flow Cytometer (BD, San Jose, CA) by Three ratios of EPIs, alone and embedded in fibrin,
acquiring at least 30 000 cells. were stained to determine the viability at three time
points, 1, 5, and 10 d. EPIs were removed from the
agarose well and rinsed three times with DPBS (Life
2.8. Transmission electron microscopy (TEM)
Technologies). A 300 μl solution of DPBS (Life
imaging
Technologies) containing 2 μM calcein-AM (Invitro-
TEM was performed using a Tecnai G2 20 microscope
gen, Carlsbad, CA) and 4 μM ethidium homodimer
(FEI Company, Hillsboro, OR). EPIs were fixed in 4%
(Life Technologies) was added to the EPIs or fibrin
paraformaldehyde (Sigma-Aldrich) for 30 min and
constructs. Plates were protected from light and
then centrifuged shortly to form cell pellet and quickly
incubated at 37 °C in 5% CO2 for up to 3 h followed by
washed in 0.1 M cacodylate buffer. The pellet was then
subjected to 1% OsO4 treatment for 60 min. After the rinsing with DBPS three times. Free-standing EPIs and
OsO4 treatment, the pellet was carefully washed in the those embedded in fibrin were transferred to the glass-
cacodylate buffer again for 10 min. Next, En Bloc bottom dishes for imaging on confocal laser scanning
staining was carried out using 2% uranyl acetate microscopy (Olympus FV10i, Olympus, America Inc.,
diluted in 50% ethanol for 30 min. Samples were then Center Valley, PA) to detect calcein (excitation 499,
dehydrated using graded ethanol solutions (50%– emission 520) and ethidium homodimer (excitation
95%) on ice and in propylene oxide before finally 577, emission 603). Ten representative areas of each
embedding them in epoxy resin. Following the epoxy sample type, ratio, and time point were randomly
embedding, 70 nm sections of the specimen were selected for imaging. Viability was quantified using
obtained by ultramicrotomy (UC6, Leica EM) fol- ImageJ (NIH). In total, a minimum of 180 EPIs from
lowed by mounting them on TEM grids. The grids three separate runs were quantified.
were stained post-sectioning with uranyl acetate and Cell apoptosis was determined by caspase-3/7
lead citrate to increase the contrast and observe activity assay at three time points (Day 1, 5, and 10).
cellular level details. EPIs were transferred from the agarose well to a tube,
The TEM images of free-standing EPIs were used and incubated in 1 ml fresh cell culture media with
to quantify insulin granules in βTC3-only, 2:1, and 1:1 10 μl CellEvent™ caspase-3/7 green ReadyProbes™
EPIs. The images were processed on ImageJ software reagent (Thermofisher) for 2 h at 37 °C in the dark.
(National Institutes of Health, NIH) using the Multi- Stained EPIs were observed using laser scanning
point Tool plugin to enumerate the insulin granule microscopy (Olympus FV10i).
content. Cell proliferation was determined by a 3-(4,
5-Dimethylthiazol-2-yl)-2, 5-Diphenyltetrazolium
2.9. Culturing EPIs in fibrin hydrogel constructs Bromide (MTT) assay (Life Technologies). EPIs from
Fibrin hydrogel was prepared by blending fibrinogen one complete mold were used per each time point for
protein isolated from bovine plasma (Sigma-Aldrich) each cell ratio. An equivalent number of cells not cul-
and bovine thrombin from plasma (Sigma-Aldrich). tured in the molds was used as a reference control.
Both solutions were dissolved separately in Dulbecco’s EPIs were flushed from agarose wells, washed twice
phosphate buffered saline (DPBS) in the following with DPBS (Life Technologies), and suspended in
concentration: 10 mg ml−1 fibrinogen and 3 U ml−1 100 μl of the media without phenol red. To each sam-
thrombin at 37 °C. Both components were combined ple, 10 μl of 12 mM MTT solution was added; an assay
in equal amounts yielding a final concentration of blank consisted of cell medium without cells. All sam-
5 mg ml−1 fibrinogen and 1.5 U ml−1 thrombin. EPIs ples and controls were incubated at 37 °C for 4 h. All
were gently suspended in thrombin, which was but 50 μl of the reaction solution was removed and
blended with fibrinogen. After gentle, thorough pipet- 100 μl of dimethylosulfoxide (DMSO; Sigma-Aldrich)
ting, the pre-crosslinked suspension was deposited on was added, thoroughly mixed with and incubated at
12 mm round cover slips placed in a 24-well plate. 37 °C for 10 min. Samples were mixed again and the
After 15 min of crosslinking in the incubator, cell absorbance was read immediately at 540 nm on a
media was deposited on top of fibrin. The culture Powerwave X-340 spectrophotometer (BioTek,
medium was formulated in 1:1 ratio of βTC3 media Winooski, VT) and the data was generated by KCju-
and EGM-2V media (Lonza) as it yielded the nior software (Biotek).

4
Biofabrication 10 (2018) 035003 M Hospodiuk et al

2.11. Immunostaining imaging and kept in −20 °C for the insulin enzyme-linked
To determine the cell distribution and demonstrate immunosorbent assay (ELISA) analysis and the
endothelial sprouting and angiogenesis, immunos- remaining cell debris was kept separately in −20 °C for
taining was performed using a rabbit anti-platelet DNA quantification. Mouse ultrasensitive insulin
endothelial cell adhesion molecule (PECAM-1) anti- ELISA kit (Alpco, Salem, NH) was used to detect insu-
body (Ab28364, Abcam, Cambridge, MA) to label lin secretion of EPIs and 2D cultures after glucose
RHMVECs and guinea pig anti-insulin antibody simulation test at the 1st, 5th, and 10th day of experi-
(Ab7842, Abcam) for βTC3 cells. Nuclei were stained ments. ELISA protocol was performed according to
with Hoechst 33258 (ThermoFisher). manufacturer’s instruction. Briefly, 5 μl of standard
The EPIs embedded within the fibrin constructs solution or sample was applied in duplicate wells of the
were cultured in a 24-well plate for 1 and 3 d, and then pre-coated ELISA microplate and then, 75 μl of HRP
fixed overnight with 4% paraformaldehyde (Sigma- conjugate was added. Plate was incubated for 2 h on a
Aldrich) at 4 °C. Constructs were then washed 3 times shaker (300 rpm) and then rinsed thoroughly six times
per 10 min in DPBS at the room temperature. Permea- with wash buffer. After careful removal of the final
bilization was performed with 0.25% Triton X-100 wash, 100 μl of TMB substrate was pipetted into each
(Sigma-Aldrich) and 5% normal goat serum (Abcam) well; the plate was incubated for 30 min on a platform
diluted in DPBS and incubated for 1 h. Primary anti- shaker protected from light. Afterward, 100 μl of stop
bodies were diluted in blocking solution (1:50 for solution was added into each well. Absorbance read-
PECAM-1 and 1:200 for insulin) applied to constructs. ings were performed at 450 nm on a Powerwave X-340
After overnight incubation at 4 °C, samples were spectrophotometer (BioTek) and insulin concentra-
washed 3 times (10 min each in DPBS). PECAM sec- tions were calculated by KCjunior software. The DNA
ondary goat anti-rabbit antibody (Alexa Fluor 647, content from the cell pellet of each group and time
Life Technologies) diluted 1:500, was incubated within point was detected by CyQUANT Cell Proliferation
the constructs for 1 h at room temperature in dark and Assay Kit (ThermoFisher) and the DNA content of
then washed for 10 min in DPBS. Then, insulin goat βTC3 cells were obtained by multiplying the total
anti-guinea pig secondary antibody (Alexa Fluor 568, DNA content with the percentage of βTC3 cells in
Life Technologies), diluted 1:500, was incubated EPIs at different ratios and time points. The obtained
within the constructs for 1 h at room temperature in DNA content for βTC3 cells was used to normalize the
dark. Staining was followed by 10 min. DPBS wash insulin quantity per 105 βTC3 cells.
and incubated for 30 min in 5 μg ml−1 Hoechst. Sam-
ples were washed three times per 10 min in DPBS, fol- 2.13. Quantification of endothelial sprouting
lowed by quick rinse in distilled water and then placed Images of EPIs encapsulated in fibrin were taken at
on a droplet of Vectashield (Vector Laboratories) on Day 1, 3 and 5 on the EVOS FL Auto. These images
glass-bottom dishes for imaging on a confocal laser were then processed on ImageJ software (NIH) using
scanning microscope (Olympus FV10i) by lasers Alexa the Angiogenesis Analyzer plugin [27] to generate a
Fluor 568 (excitation 577, emission 603) and Alexa skeleton of the sprouts. The generated skeleton was
Fluor 647 (excitation 653, emission 668). Images were further analyzed using the Analyze Skeleton plugin to
pseudo-colored, green for PECAM and red for insulin. quantify the sprout length and number. The average
sprouting length and the average sprout number was
2.12. Insulin secretion analysis calculated for a set of 10 EPIs for each group (2:1 and
The insulin secretion analysis was conducted at 3 time 1:1) at each time point (Day 1, 3 and 5).
points (1, 5, and 10 d) of βTC3-only, 2:1, and 1:1 of
βTC3 and RHMVEC, respectively. 1x Krebs buffer 2.14. Histology study
was freshly prepared (25 mM HEPES, 115 mM NaCl, EPIs in three groups (βTC3-only, 2:1, and 1:1)
24 mM NaHCO3, 5 mM KCl, 1 mM MgCl2, 0.2 g embedded in fibrin on 12 mm cover slips were
0.1% BSA dissolved MQ H2O). To the final solution cultured for five days in a 24-well plate. The EPIs-laden
was added 2.5 mM CaCl2 and the pH was adjusted to constructs were stabilized by overlaying with 300 μl of
7.4 with 1 M NaOH. This solution was filtered through 1.5% agarose (RPI Corp.), prior to fixation in 4% (v/v)
0.22 μm filter and stored at 4 °C. paraformaldehyde (Sigma-Aldrich) at 4 °C overnight.
Only a single glucose stimulation was performed The constructs were embedded in paraffin using an
due to unresponsive character of βTC3 cells on ele- automatic tissue processor (Leica TP 1020). The
vated glucose levels [26]. Therefore, 28 mM glucose constructs were gradually dehydrated in alcohol and
solution was prepared in the 1x Krebs buffer. All of the sectioned cut into 8 μm sections and placed onto
EPIs (βTC3-only, 2:1, and 1:1 groups), were shortly charged slides. Sections were stained with hematoxylin
rinsed in a glucose solution and then incubated in a and eosin (H&E) using Leica Autostainer XL (Leica).
500 μl of fresh solution for 1 h in 37 °C and 5% CO2. After H&E staining, cover slips were mounted to the
After incubation, samples were centrifuged at slides with Xylene Substitute Mountant (Thermo-
2000 rpm for 5 min, the supernatant was extracted fisher). The slides were viewed under the Olympus

5
Biofabrication 10 (2018) 035003 M Hospodiuk et al

BX51 (Olympus), and images were captured with the increased over 33% over a 10-day period. A similar
manufacturer’s software. trend was observed when βTC3 were seeded at
2×106 cells/mold and increased over 24%. The aver-
2.15. Statistical analysis age diameter of βTC3-only EPIs seeded at 3×106
Due to the large number of data points analyzed, the cells/mold increased only by 16% over the 10-day per-
average diameter of EPIs is shown as the mean, and iod suggesting that the micro-well size limits the max-
error bars represent the standard deviation from the imum diameter of the EPIs. Similar results were seen
mean. All other data are reported as the mean with in EPIs formed by 2:1 and 1:1 ratio. When 1×106
error bars indicating the standard error of the mean. cells/mold were seeded, the average diameter in 2:1
Statistical significance was determined using one-way and 1:1 group increased by 25% and 15%, respectively.
analysis of variance on MINITAB 17.3 (Minitab Inc., Using 2×106 cells/mold, the average EPI diameter
State College, PA). Post-hoc Tukey’s multiple compar- increased by 15% for both 2:1 and 1:1 groups over the
ison test was used to determine the individual 10-day period. Finally, cell seeded at the density of
differences among the groups. Results were considered 3×106 cells/mold increased by 12% (2:1) and 9%
significant with a confidence level of 95%, where (1:1) over the 10-day period. Based on growth rate and
p<0.05 (*), p<0.01 (**), and p<0.001 (***). the necessity to maintain an average diameter below
300 μm, 2×106 cells/mold was used for the rest of
the study.
3. Results SEM images on three days-cultured EPIs demon-
strated solid, compact and spherical-shaped structure
3.1. EPI fabrication and characterization
(figure 2(B)). The ECM was deposited by cells and pre-
In this investigation, βTC3 cells and RHMVECs were
sented as slightly irregular crown on the surface, which
co-cultured for EPIs formation in three ratios, βTC3-
is magnified in the bottom row of figure 2(B).
only, 2:1 and 1:1, for βTC3 cells and RHMVECs,
RHMVECs contributed directly to synthesizing the
respectively. In EPIs, βTC3 cells provided the insulin-
ECM, which includes collagen type IV and laminin
secreting component and RHMVECs served to
[29] and serves as physical barrier for immune system
strengthen EPI formation and facilitate angiogenic
cells. Therefore, in 2:1 and 1:1 groups, higher amount
sprouting.
of ECM was deposited and EPI in these groups were
Agarose has been extensively used for molds that
characterized by smoother surface topology and
facilitate cell fusion in 3D [28]. The printed plastic
round morphology.
mold (figure 1(A)) is characterized by high-resolution
The flow cytometry results show two distinguish
of fine details and a superior surface finish. When filled
populations of tdTomato+ RHMVECs and βTC3 at
with liquefied agarose, the mold formed reproducible
every time point with some unstained cell debris
uniform hydrogel with 124 micro-wells with a 300 μm
(figure 3). At Day 1, the detected ratio was consistent
diameter, as shown in figure 1(B) (top). After 24 h of
with the experimental setup, where 2:1 group con-
incubation in a biocompatible, non-toxic, mechani-
tained twice more βTC3 cells than RHMVECs and in
cally stable, and non-adhesive agarose mold, EPIs had
1:1 ratio the proportion was equal. The βTC3 ratio in
a compact and rigid morphology (figure 1(B), bottom
EPIs diminished from 54% to 23% and 37% to 11%
left and right) that allowed to be flushed out of the
between Day 1 and 10 for 2:1 and 1:1 group, respec-
mold with a gentle pipetting. Surface tension for each
tively. On the other hand, RMHVEC ratio increased
EPI group was measured at Days 1 and 5. On Day 1,
surface tensions of βTC3-only, 2:1, and 1:1 groups from 26% to 46% and 38% to 66% between Day 1 and
were measured at 2.16±0.56, 5.99±2.09, and 10 for 2:1 and 1:1 group, respectively.
19.99±4.79 mN m−1, respectively. The surface ten-
sion of 1:1 group EPI was found to be significantly 3.2. Cell viability and proliferation
greater than either βTC3-only or 2:1 ratio EPI at Day Cell viability in EPIs was detected over a 10-day
1. However, there was no noteworthy difference culture, for both free-standing EPIs and EPIs
between βTC3-only and 2:1 ratio. On Day 5, the sur- embedded in fibrin hydrogel (with sprouting angio-
face tension of 2:1 and 1:1 EPIs were determined to be genesis). The average viability of free-standing βTC3-
12.08±6.65 and 17.08±3.83 mN m−1. At Day 10, only EPIs was 87.41%±4.24% at Day 1,
the surface tension of 2:1 and 1:1 groups were 73.25%±1.02% at Day 5, and 56.44%±5.09% at
3.84±2.06 and 13.25±6.47 mN m−1, respectively. Day 10 (figure 4(A)). The average viability decreased
The surface tension of EPI in 1:1 group was sig- significantly (by over 35%) over the 10-day period.
nificantly greater than that of 2:1. The surface tension This trend was not observed for the 2:1 and 1:1 groups,
of βTC3-only group was not able to be measured at where cell viability was maintained from 83% to 91%
Day 5 and 10, due to their weak structural properties. for 2:1 group and 88% to 92% for 1:1 group during the
EPI diameters were measured at 1, 5, and 10 d 10-day culture. The viability rate did not change
(figure 2(A)). The average diameter of βTC3-only significantly neither for 2:1 nor 1:1 group over the 10-
group seeded at concentration 1×106 cells/mold day period. Strongly decreasing tendency in viability in

6
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Figure 2. (A) EPI diameter changed over time for three different groups (n=15). (B) SEM images of three-day cultured EPIs with a
high-magnification view of their surface morphology.

βTC3-only group suggest that the presence of EPIs (right panel), where βTC3-only group exhibited
RHMVECs increased the viability of EPIs. The viabi- significant cell death. Based on the results of caspase-3
lity study revealed similar tendencies for EPIs assay, more apoptosis presented in βTC3-only EPI in
embedded in fibrin (figure 4(A), right), where similar both free-standing and fibrin-embedded groups.
tendencies were noted. The average viability of βTC3- These results were in line with the viability study
only group was 86.09%±4.38% at Day 1, presented in figure 5.
75.58%±7.14% at Day 5, and 58.19%±4.81% at
Day 10. The average viability decreased by 32.41% 3.3. Functional analysis of EPIs
over the 10-day period. Such trend was not observed In TEM images, insulin granules were visible in 70 nm
for other groups, where the average viability was thick sections of EPIs. These unique β-cell granules
remained ∼85% for 2:1 group and ∼90% for 1:1 group were visible in different stages of maturation and were
over time. The MTT results (figure 4(B)) show that surrounded by characteristic halo and membrane
proliferation of βTC3-only EPIs significantly [30]. The granules were present in each group
decreased over time by more than 60% from Day 1 to (figure 6(A)) in both early and late culture stages. Early
10. The 2:1 and 1:1 ratio EPIs had a stable proliferation insulin granules had larger, sparse granules with faded
rate (which did not differ significantly over time) membrane (figure 6(A), red arrows); mature granules
indicating that 3D co-culture of βTC3 and RMHVEC had a dense core and clearly defined membrane
supported cell proliferation and confirms the live/ (figure 6(A), blue arrows). The insulin granules of
dead staining results. The representative pictures from βTC3 cells, which is a mouse insulinoma cell line, had
live/dead staining are presented in figure 4(C), for a diameter not exceeding 250 nm that corresponds to
both free-standing (left panel) and fibrin-embedded the size of rat insulin granules (243 nm) reported by

7
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Figure 3. Flow cytometry based on segregated EPIs made of tdTomato+ RHMVECs and βTC3 cells stained with insulin. The analysis
was performed at Days 1, 5, and 10 for both groups, 2:1 and 1:1.

Fava et al [30]. The number of insulin granules in free- and 2). When placed close to each other, EPIs were
standing 2:1 and 1:1 ratio EPIs after three days of able to fuse, contract and create a void space in fibrin
culture was slightly higher than βTC3-only group, but matrix as shown in figure 7(A) (red arrows). Fibrin
not statistically significant (figure 6(B)). microstructure allowed migration of RHMVECs,
Free-standing EPIs in two ratios, 2:1 and 1:1, were while βTC3 cells maintained their position within
immunostained for insulin to label the βTC3 cells, and the EPIs.
PECAM-1 to identify the ECs. The confocal images, Immunocytochemistry was performed on EPIs to
shown in figure 6(C), indicate uniform distribution of understand the sprouting behavior of RHMVECs.
each cell type and strong expression of insulin in EPIs. RHMVECs exhibited typical endothelial sprouting
Angiogenesis was not observed within free-stand- into the fibrin matrix, while βTC3 cells (stained with
ing EPIs. insulin) were maintained within the EPI over the cul-
The insulin secretion in βTC3-only EPIs was ture period. When EPIs were placed close together,
maintained at the same level over 10 d, around 3.6 pg sprouts between them tended to merge as what
insulin/105 cells, while the 2:1 and 1:1 EPIs represent appears to be a nascent vascular network (figure 7(B),
statistically significant, four-fold higher insulin secre- top). Also, RHMVECs started to form capillaries after
tion per cell at Day 1, 14.4 and 15.3 pg insulin/105 two days in culture (figure 7(B) (middle) and supple-
cells, respectively. On Day 5, higher levels of insulin mentary video 3) and created angiogenic sprouts over
were detected in 2:1 and 1:1 groups compared to the a five-day time period (figure 7(B), bottom).
βTC3-only group, with 16.4 and 7.8 versus 3.6 pg As supportive cells (such as fibroblasts, smooth
insulin per 105 cells, respectively. On Day 10, βTC3- muscle cells or pericytes [31]) were not present in
only group secreted 3.9 pg insulin/105 cells, sig- fibrin matrix, intensive capillarization was formed
nificantly lower than 2:1 (with 11.1 pg/105 cells) and around EPIs at Day 10 disabling the quantification in
1:1 (18.5 pg/105 cells) groups. ImageJ. Moreover, observation at a mid-time-point
was essential to understand the sprouting mechanism.
3.4. Sprouting angiogenesis Therefore, Days 1, 3 and 5 were preferred as time
In this work, EPIs were embedded in fibrin hydrogel to points instead of Days 1, 5 and 10. Average sprouting
induce angiogenesis. Over the 64 h culture, EPIs were length and the number of sprouts were determined for
observed to form extensive capillarization 2:1 and 1:1 groups at three time points (1, 3, and 5 d) as
(figure 7(A)). Endothelial sprouts developed approxi- shown in figure 8(A). For 2:1 group, sprouting length
mately 20 h after seeding and formed a more complex increased by 28% by Day 3 and 64% by Day 5. The
vasculature over time. Complete time lapse images are number of sprouts increased by 75% from Day 1 to
provided in the supplementary data section (Videos 1 Day 3, but no significant increase was observed from

8
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Figure 4. (A) Quantification of cell viability on free-standing EPIs (left) and EPIs embedded in fibrin (right) over 1, 5, and, 10 d
(n = 10). (B) Proliferation rate conducted on MTT assay over the same time points (n=4). (C) Representative pictures of live/dead
staining at Day 1 (top row), 5 (middle row), and 10 (bottom row) for free-standing (left panel) and fibrin-embedded (right panel)
culture for βTC3-only (left), 2:1 (middle), and 1:1 (right) group of EPIs.

Day 3 to Day 5. A similar increase in the average sprout ability to fabricate a large number of EPIs (in this case,
length was also observed for 1:1 group, which was 124) at one time. The shortcomings of this technique
increased by 65% from Day 1 to Day 3 and 83% from are random distribution of cells, especially for co-
Day 1 to Day 5. cultured cells, a variation in the exact cell number per
In order to investigate the micro-architecture for EPI, and the necessity to flush out cells where the
capillarization, EPIs of each ratio were processed for physical force can trigger off disaggregation. The size
histological sectioning and H&E staining. The morph- of wells affected the EPIs formation. In our prelimin-
ology in both 2:1 and 1:1 groups showed the presence ary study, molds with well sizes of 500 and 800 μm
of channel-like structures within the EPI. In 2:1 group, were tested (supplementary figure 2), where EPIs
where there were more βTC3 cells than RHMVECs, overgrew and large deviations in their diameters were
the morphology was more compact, with smaller observed whereas molds with 300 μm well diameter
sprouts (figure 8(B), middle red arrows). The 1:1 yielded EPIs with tight control on their dimensions.
group is characterized by looser arrangement with lar- Free-standing βTC3-only EPIs were very fragile after a
ger channel-like structures. More pronounced con- few days in culture. They disaggregated easily, particu-
traction of EPI at the 1:1 ratio caused a tear in the larly during the extraction from the mold. βTC3 cells
fibrin hydrogel, as shown in figure 8(B) (right, blue were unable to maintain a ∼300 μm diameter due to
arrows). minimal ECM deposition [15]. These observations
were validated by surface tension measurements,
where βTC3-only EPI demonstrated low surface
4. Discussions tension on Day 1, which was not even detectable by
Day 5. This can be attributed to limited ECM deposi-
Formation of tissue spheroids in molds made of tion by βTC3 cells as well as the hypoxia-induced cell
agarose [24, 32, 33], polyethylene glycol [34] and death as presented in the viability study. The 2:1 and
polydimethylsiloxane [35] has been previously dis- 1:1 groups demonstrated significantly higher surface
cussed elsewhere. This affordable technique offers the tensions with better structural integrity that did not

9
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Figure 5. Representative pictures of caspase-activated apoptosis at 1 (top row), 5 (middle row), and 10 (bottom row) days of free-
standing (left panel) and fibrin-embedded (right panel) culture for βTC3-only (left), 2:1 (middle), and 1:1 (right) ratio of EPIs.

Figure 6. (A) Ultra-morphology of EPI showing the vesicles containing insulin granules in each group: βTC3-only (left), 2:1 (middle),
and 1:1 (right). The arrows indicate immature (red) and mature (blue) insulin granules. (B) Number of insulin granules per cell in
βTC3-only, 2:1, and 1:1 groups (n=15). (C) Immunocytochemistry of EPIs with RHMVECs stained for PECAM (green), βTC3
insulin (red), and nuclei (blue). (D) Insulin secretion in picograms normalized per 105 βTC3s in βTC3-only, 2:1, and 1:1 groups in
respect to 1, 5, and 10 days of culture (n=6).

change from Day 1 to 5. This is likely due to the 1:1 groups led to a negative effect on sprouting
deposition of ECM proteins by ECs [15], and can angiogenesis as robust capillary formation did not take
readily be seen in the SEM images presented in place in mature EPIs during the experiments (data not
figure 2(B). In EPIs, intercellular adhesion forces shown) confirming on earlier study [37]. Therefore,
determined the final configuration of the spheroid by EPIs were embedded in fibrin after the overnight
minimizing the total surface free energy. As a result, culture in molds.
EPIs with RHMVECs became more spherical in shape The size of EPIs in each group increased over time,
[36]. The increased mechanical properties and higher even as the viability decreased. We assume it was
amount of deposited and remodeled ECM in 2:1 and caused by proliferation zone in which the outside layer

10
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Figure 7. EPIs seeded within fibrin hydrogel. (A) Time lapse images of 2:1 (top) and 1:1 (bottom) groups over a 64 h period (note: the
tear caused by EPI contraction is shown by red arrows). (B) Immunocytochemistry images illustrating endothelial cells sprouting.
Sprouting between two EPIs in 2:1 group (top) and in the core and outside of the EPI in 1:1 group (middle and bottom).
Immunostaining with PECAM (CD31) stains RHMVECs in green, insulin stains βTC3 in red, and nuclei are shown in blue.

thickness increased despite the necrotic core in the The flow cytometry was performed to detect the
βTC3-only group [38]. The growth rate was stable as population of βTC3 cells and RHMVECs over the 10-
the EPIs were maintained in micro-wells, which day culture for both ratios (figure 3). Results con-
restrained their proliferation. In the literature, it was firmed the original seeding ratios at Day 1; however, at
reported that necrosis of spheroids in the core begins the later time points, RHMVECs presented a tendency
to occur when the diameter of the structure exceeds to proliferate in a higher rate than βTC3 cells, causing
150 μm [38]. Necrosis is caused by the limited micro- outgrowth over the 10-day culture for both ratios
well volume and therefore the lack of nutrient pene- (figure 3). Thus, further research is needed to investi-
tration to the EPI core. In 2:1 and 1:1 groups, the gate a gold ratio between β-cells and ECs to maintain
strong adhesion exhibited by RHMVECs and the ECM high viability, but avoiding overgrow of ECs among
components produced by them could contribute to EPIs in a long-term culture.
the limited expansion of the EPIs. However, EPIs During culture, EPIs were surrounded with fresh
exhibited an increase in their diameter under all seed- media within immediate. Viability of βTC3-only
ing conditions, suggesting cell proliferation, proper group decreased significantly over 10 days, while the
growth conditions and a synergistic relationship 2:1 and 1:1 ratio EPIs maintained their viability
between the pancreatic and vascular cells. (∼85%) in same conditions. While it was expected that

11
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Figure 8. (A) Average sprouting length (left) and the number of sprouts (right) over a period of five days (n=5). (B) H&E staining of
EPIs; βTC3-only (left), 2:1 (middle), and 1:1 (right) cultured in fibrin and imaged at 20× and 40×. The blue arrows indicate the void
space caused by contraction of EPIs and the red arrows indicate the endothelial sprouting.

2:1 and 1:1 groups would be denser than βTC3-only growth of RHMVECs and capillarization. We can
one, we did not observe a significant decrease in their speculate that micro-capillaries could be instrumental
viability. Since free-standing EPIs did not exhibit in delivering nutrients to the core of the EPI, enhan-
angiogenesis, the phenomena on sustained viability cing the viability of βTC3. We sometimes observed
could be explained by the research of Korff and Augus- EPIs with a dead core. Since live/dead staining is not
tin [39], which suggests that while the single sus- specific for the cell lines, we could not distinguish the
pended cells cannot survive, the aggregated ECs death rate between βTC3 cells and RHMVECs; how-
mediate intercellular contact and contribute the cell ever, weak fluorescence signal from insulin in the core
survival. The external layer of ECs express polarized indicates that more death βTC3 cells were present in
surface molecules that lead to contact development the core compared to the outer regions. Such weak
between cells. In the core, RHMVECs were exposed to insulin signal did not appear in EPIs with smaller dia-
necessary survival factors, which prevented apoptosis. meter even in longer incubation periods as shown in
Additionally, we suspect that βTC3 cells secrete nitric supplementary figure 3.
oxide and VEGF that additionally supported ECs sur- The immature and mature granules differ in inter-
vival and RHMVECs increased the mechanical prop- nal composition and organization. The immature
erties of the EPIs giving the proper matrix to grow for insulin granules, visible in the TEM images
βTC3 cells [12]. (figure 6(A)), underwent a maturation process, where
For EPIs embedded in fibrin, the media was added proteolytic conversion began from proinsulin to insu-
on the top of constructs and therefore cells had limited lin in the granule core. The process is called a passive
exposure to media. The decrease in the viability of condensation [40], where insulin is crystallized in the
βTC3-only group within 10 d (figures 4–5) was similar form of granules with zinc and calcium elements in a
when they were cultured in free-standing form. We dense core. Additionally, the uncondensed proteins
can speculate that 3D matrix formed by fibrin are removed outside the membrane [41]. Also, the
mechanically supported βTC3 cells to maintain their membrane undergoes selective trafficking to one of
viability with limited supplementation of media com- two pathways, secretion or degradation. Both matura-
pared to the free-standing group. EPIs in 2:1 and 1:1 tion states were present for each EPI group indicating
ratio embedded in fibrin maintained a high viability, the natural process of insulin production. The free-
similar to the free-standing ones. The viability in fibrin standing EPIs were processed for TEM imagining on
was maintained at a high level due to the expansive Day 3 of culture. Slightly higher insulin granule

12
Biofabrication 10 (2018) 035003 M Hospodiuk et al

content was detected for 2:1 and 1:1 groups compared by steric hindrance and the availability of fibrin matrix
to βTC3-only; however, no significant difference was for tube formation. The VEGF factor, contained in the
found. The immunostaining on the 2nd day media and secreted by βTC3 cells, promoted ECs
(figure 6(C)) presented a uniform distribution of both motility, proliferation, and filopodia extension. Toge-
cell types with strong expression of insulin within ther with the Notch signaling, the capillary lead tips
the EPIs. were recruited and controlled as present in figure 7(A)
The normalized results of insulin secretion over [53]. Also, local suppression of ECs motility and for-
time in both 2:1 and 1:1 groups indicate increased mation of new intra-cell junctions resulted in connec-
insulin secretion by βTC3 cells significantly after 10 tions into tube-like capillary sprouts (figure 8(B)). The
days of culture compared to the βTC3-only group as histology images revealed the formation of channel-
shown in figure 6(D). The βTC3-only group con- like structures. We suspect that RHMVECs provided
tinued to secrete the same amount of insulin calcu- an enhanced flow of nutrients to the core of EPIs as
lated per 100 k live cells, even if the average viability well as increased mechanical strength to the structure
decreased. This indicates that the presence of via the deposition of ECM.
RHMVECs improved the amount of insulin produced A critical step in successful clinical transplantation
by βTC3 cells and favored the functionality of EPIs. of the pancreas is the revascularization of the organ
EPIs in 2:1 and 1:1 groups started to form capil- [7, 54]. The interaction between ECs and pancreatic
laries within several hours after embedding in fibrin. If islet cells is crucial to initiate growth, maturation and
they were placed close to one another, a significant function to the endocrine system. It has been reported
contraction was observed. The reason behind the cel- that vascularization is critical issue after transplanta-
lular contraction plays significant role in the assembly tion as it usually diminishes over time [8, 55, 56]. As
process as explained elsewhere [42–45]. Kinetics demonstrated in this investigation, it is possible to
increases by changes in morphology and tissue stabi- recapitulate the synergistic relationship between ECs
lity during self-assembly [42]. Since the surface ten- and pancreatic β-cells to create a functional 3D in vitro
sion and the mechanical contraction of fusing EPIs model of pancreatic endocrine tissue.
was stronger than the stiffness of surrounding fibrin, it
might cause damage in fibrin matrix as shown in
5. Conclusions
figure 7(A) (pointed by red arrows). The fibrin hydro-
gel particularly serve capillarization and has been
Engineering pseudo islets with capillarization is criti-
documented by a wide range of in vitro models of both
cal, since the in-and-out flow of intra-islet nutrients as
angio- and vasculogenesis [46–48]. Fibrin is naturally
well as maintenance of the crosstalk between β-cells
angiogenic, promotes cellular production of ECM
and EC is necessary for proper islet function in vivo.
proteins for achieving stable microvascular network
Here, we report the sprouting angiogenesis in β-cells
for creation of angiogenic models [49]. We observed
clusters (EPIs) in 3D and demonstrated its role in
that capillarization was formed in the bodies of EPI,
improving the viability and functionality of clusters
and also was directed to surrounding (figure 7(B)).
over time. Functional EPIs can act as building blocks
ECs associated into capillaries were interconnected
to form larger, viable tissue constructs for various
with one another forming a branched structure. This
purposes such as organs for transplantation or disease
behavior has been observed in a number of studies [47,
models for early phase pharmaceutical testing or
50–52]. The RHMVEC sprouting length for both
research investigations for complex, multi-scale vas-
ratios, 2:1 and 1:1, showed similar increasing tendency
cularization and multicellular interactions. As a future
over five days (figure 8(A), left). The higher number of
work, EPIs generated in this study will be bioprinted
sprouts at Day 1 in 2:1 group compared to 1:1 group
into a perfusable vascular bed in a larger scale for
might be caused by significantly higher surface tension
fabrication of pancreas-on-a-chip devices for pharma-
or the VEGF production by βTC3 cells; however, on
ceutical testing of T1D-related drugs.
Day 3, both ratios present similar average sprouting
length. The number of sprouts did not differ sig-
nificantly between 2:1 and 1:1 groups (figure 8(A), Acknowledgments
right). The results indicate that EPIs sprouting length
was similar for 2:1 and 1:1 groups at Days 3 and 5. We This work has been supported by National Science
found that interesting, since 1:1 group was loaded with Foundation (Award # 1624515), Diabetes in Action
more RHMVECs, we assume that RHMVECs attained Research and Education Foundation and the ENGINE
maximum sprouting length capabilities in both ratios grant from the Penn State University. The authors are
regardless of the EC population. Due to RHMVECs grateful to the support from the Turkish Ministry of
crowding around the EPIs at Day 5, most of the National Education for providing graduate scholar-
sprouts seemed to fuse and form thicker capillaries. ship to BA. The authors are thankful to the Huck
Therefore, the quantified number of sprouts was simi- Institute of Life Sciences and the Materials Research
lar as compared to Day 3. As the number of sprouts Institute for their facility and staff support for imaging
increased, the growth of new sprouts was likely limited experiments. The authors also thank Drs. Adil

13
Biofabrication 10 (2018) 035003 M Hospodiuk et al

Akkouch and Ning Pan from The University of Iowa human embryonic stem cell derived pancreatic progenitor cells
in their assistance with earlier feasibility experiments Tissue Eng. A 0 1–41
[19] Johansson Å, Lau J, Sandberg M, Borg L A H,
for fabrication of cell aggregates. The authors confirm
Magnusson P U and Carlsson P O 2009 Endothelial cell
that there are no known conflicts of interest associated signalling supports pancreatic beta cell function in the rat
with this publication and there has been no significant Diabetologia 52 2385–94
financial support for this work that could have influenced [20] Sabra G and Vermette P 2013 A 3D cell culture system:
separation distance between INS-1 cell and endothelial cell
its outcome.
monolayers co-cultured in fibrin influences INS-1 cells insulin
secretion Biotechnol. Bioeng. 110 619–27
[21] Paget M B, Murray H E, Bailey C J, Flatt P R and Downing R
ORCID iDs 2011 Rotational co-culture of clonal β-cells with endothelial
cells: effect of PPAR-γ agonism in vitro on insulin and VEGF
Ibrahim T Ozbolat https://orcid.org/0000-0001- secretion Diabetes, Obesity Metab. 13 662–8
8328-4528 [22] Penko D, Mohanasundaram D, Sen S, Drogemuller C, Mee C,
Bonder C S, Coates P T H and Jessup C F 2011 Incorporation of
endothelial progenitor cells into mosaic pseudoislets Islets 3
References 73–9
[23] Spelios M G, Kenna L A, Wall B and Akirav E M 2013 In vitro
[1] Atkinson M A, Eisenbarth G S and Michels A W 2014 Type 1 formation of β Cell pseudoislets using islet-derived endothelial
diabetes Lancet 383 69–82 cells PLoS One 8 1–7
[2] Cnop M, Welsh N, Jonas J C, Jorns A, Lenzen S and Eizirik D L [24] Napolitano A P, Dean D M, Man A J, Youssef J, Ho D N,
2005 Mechanisms of pancreatic beta-cell death in type 1 and Rago A P, Lech M P and Morgan J R 2007 Scaffold-free three-
type 2 diabetes: many differences, few similarities Diabetes 54 dimensional cell culture utilizing micromolded nonadhesive
S97–107 hydrogels Biotechniques 43 494–500
[3] Faglia E, Clerici G, Clerissi J, Gabrielli L, Losa S, Mantero M, [25] Guevorkian K, Colbert M J, Durth M, Dufour S and
Caminiti M, Curci V, Lupattelli T and Morabito A 2006 Early Brochard-Wyart F 2010 Aspiration of biological viscoelastic
and five-year amputation and survival rate of diabetic patients drops Phys. Rev. Lett. 104 1–4
with critical limb ischemia: data of a cohort study of 564 [26] Skelin M, Rupnik M and Cencic A 2010 Pancreatic beta cell
patients Eur. J. Vascular Endovascular Surg. 32 484–90 lines and their applications in diabetes mellitus research
[4] Leksell J K, Wikblad K F and Sandberg G E 2005 Sense of ALTEX 27 105–13
coherence and power among people with blindness caused by [27] Woo S, Mun S, Lee M, Al-hilal T A, Kyung C, Kon J, Kim I,
diabetes Diabetes Res. Clin. Pract. 67 124–9 Yoon S and Byun Y 2015 LHT7, a chemically modified
[5] Forbes J M and Cooper M E 2013 Mechanisms of diabetic heparin, inhibits multiple stages of angiogenesis by blocking
complications Physiol. Rev. 93 137–88 VEGF, FGF2 and PDGF-B signaling pathways Biomaterials 37
[6] Migliorini A, Bader E and Lickert H 2014 Islet cell plasticity 271–8
and regeneration Mol. Metab. 3 268–74 [28] Mehesz A N, Brown J, Hajdu Z, Beaver W, da Silva J V L,
[7] Jansson L and Carlsson P-O 2002 Graft vascular function after Visconti R P, Markwald R R and Mironov V 2011 Scalable
transplantation of pancreatic islets Diabetologia 45 749–63 robotic biofabrication of tissue spheroids Biofabrication 3
[8] Carlsson P O, Palm F and Mattsson G 2002 Low 25002
revascularization of experimentally transplanted human [29] Godoy P et al 2013 Recent advances in 2D and 3D in vitro
pancreatic islets J. Clin. Endocrinol. Metab. 87 5418–23 systems using primary hepatocytes, alternative hepatocyte
[9] Ravnic D J, Leberfinger A N, Koduru S V, Hospodiuk M, sources and non-parenchymal liver cells and their use in
Moncal K K, Datta P, Dey M, Rizk E and Ozbolat I T 2017 investigating mechanisms of hepatotoxicity, cell signaling and
Transplantation of bioprinted tissues and organs: technical ADME Arch. Toxicol. 87 1315–530
and clinical challenges and future perspectives Ann. Surg. 266 [30] Fava E, Dehghany J, Ouwendijk J, Muller A, Niederlein A,
48–58 Verkade P, Meyer-Hermann M and Solimena M 2012 Novel
[10] Cleaver O and Dor Y 2012 Vascular instruction of pancreas standards in the measurement of rat insulin granules
development Development 139 2833–43 combining electron microscopy, high-content image analysis
[11] Stendahl J C, Kaufman D B and Stupp S I 2009 Extracellular and in silico modelling Diabetologia 55 1013–23
matrix in pancreatic islets: relevance to scaffold design and [31] Whisler J A, Chen M B and Kamm R D 2014 Control of
transplantation Cell Transplant. 18 1–12 perfusable microvascular network morphology using a
[12] Peiris H, Bonder C S, Coates P T H, Keating D J and Jessup C F multiculture microfluidic system Tissue Eng. C 20 543–52
2014 The B-cell/EC axis: how do islet cells talk to each other? [32] Krotz S P, Robins J C, Ferruccio T M, Moore R, Steinhoff M M,
Diabetes 63 3–11 Morgan J R and Carson S 2010 In vitro maturation of oocytes
[13] Jain N and Lee E J 2016 Islet endothelial cells derived from via the pre-fabricated self-assembled artificial human ovary
mouse embryonic stem cells Cell Transplant. 25 97–108 J. Assist. Reprod. Genet. 27 743–50
[14] Reinert R B et al 2013 Vascular endothelial growth factor-a and [33] Owens C M, Marga F, Forgacs G and Heesch C M 2013
islet vascularization are necessary in developing, but not adult, Biofabrication and testing of a fully cellular nerve graft
pancreatic islets Diabetes 62 4154–64 Biofabrication 5 45007
[15] Nikolova G et al 2006 The vascular basement membrane: a [34] Bernard A B, Lin C-C, Anseth K S, Ph D, Anseth K S and Ph D
niche for insulin gene expression and β cell proliferation Dev. 2012 A microwell cell culture platform for the aggregation of
Cell 10 397–405 pancreatic β-cells Tissue Eng. C 18 583–92
[16] Millman J R, Xie C, Van Dervort A, Gürtler M, [35] Kusamori K, Nishikawa M, Mizuno N, Nishikawa T,
Pagliuca F W and Melton D A 2016 Generation of stem cell- Masuzawa A, Shimizu K, Konishi S, Takahashi Y and
derived β-cells from patients with type 1 diabetes Nat. Takakura Y 2014 Transplantation of insulin-secreting
Commun. 7 11463 multicellular spheroids for the treatment of type 1 diabetes in
[17] Pagliuca F W, Millman J R, Gürtler M, Segel M, Van Dervort A, mice J. Control. Release 173 119–24
Ryu J H, Peterson Q P, Greiner D and Melton D A 2014 [36] Foty R A, Pfleger C M, Forgacs G and Steinberg M S 1996
Generation of functional human pancreatic β Cells in vitro Cell Surface tensions of embryonic tissues predict their mutual
159 428–39 envelopment behavior Development 122 1611–20
[18] Jaramillo M, Mathew S, Mamiya H, Goh S K and Banerjee I (PMID:8625847)
2014 Endothelial cells mediate islet-specific maturation of

14
Biofabrication 10 (2018) 035003 M Hospodiuk et al

[37] Ehsan S M, Welch-Reardon K M, Waterman M L, specifically requires the activity of membrane-type-matrix


Hughes C C W and George S C 2014 A three-dimensional metalloproteinases (MT-MMPs) J. Cell Sci. 115 3427–38
in vitro model of tumor cell intravasation Integr. Biol. Quant. (PMID:12154073)
Biosci. Nano Macro. 6 603–10 [48] Nakatsu M N, Sainson R C A, Aoto J N, Taylor K L,
[38] Mehta G, Hsiao A Y, Ingram M, Luker G D and Takayama S Aitkenhead M, Perez-del-Pulgar S, Carpenter P M and
2012 Opportunities and challenges for use of tumor spheroids Hughes C C W 2003 Angiogenic sprouting and capillary lumen
as models to test drug delivery and efficacy J. Control. Release formation modeled by human umbilical vein endothelial cells
164 192–204 (HUVEC) in fibrin gels: the role of fibroblasts and
[39] Korff T and Augustin H G 1998 Integration of endothelial cells Angiopoietin-1 Microvascular Res. 66 102–12
in multicellular spheroids prevents apoptosis and induces [49] Morin K T and Tranquillo R T 2013 In vitro models of
differentiation J. Cell Biol. 143 1341–52 angiogenesis and vasculogenesis in fibrin gel Exp. Cell Res. 319
[40] Huang X F and Arvan P 1994 Formation of the insulin- 2409–17
containing secretory granule core occurs within immature [50] Montaño I, Schiestl C, Schneider J, Pontiggia L, Luginbühl J,
B-granules J. Biol. Chem. 269 20838–44 (PMID:8063699) Biedermann T, Böttcher-Haberzeth S, Braziulis E,
[41] Hou J C, Min L and Pessin J E 2009 Insulin Granule Biogenesis, Meuli M and Reichmann E 2010 Formation of human
Trafficking and Exocytosis 1st edn (Amsterdam: Elsevier) ch 16 capillaries in vitro: the engineering of prevascularized matrices
(https://doi.org/10.1016/S0083-6729(08)00616-X) Tissue Eng. A 16 269–82
[42] Dean D M, Napolitano A P, Youssef J and Morgan J R 2007 [51] Bauer A L, Jackson T L and Jiang Y 2007 A cell-based model
Rods, tori, and honeycombs: the directed self-assembly of exhibiting branching and anastomosis during tumor-induced
microtissues with prescribed microscale geometries FASEB J. angiogenesis Biophys. J. 92 3105–21
21 4005–12 [52] Tiruvannamalai Annamalai R et al 2016 Vascular network
[43] Asghar W, El Assal R, Shafiee H, Pitteri S, Paulmurugan R and formation by human microvascular endothelial cells in
Demirci U 2015 Engineering cancer microenvironments for modular fibrin microtissues ACS Biomater. Sci. Eng. 2 1914–25
in vitro 3-D tumor models Mater. Today 0 1–15 [53] Eilken H M and Adams R H 2010 Dynamics of endothelial cell
[44] Mattix B M, Olsen T R, Casco M, Reese L, Poole J T, Zhang J, behavior in sprouting angiogenesis Curr. Opin. Cell Biol. 22
Visconti R P, Simionescu A, Simionescu D T and Alexis F 2014 617–25
Janus magnetic cellular spheroids for vascular tissue [54] Carlsson P O, Berne C and Jansson L 1998 Angiotensin II and
engineering Biomaterials 35 949–60 the endocrine pancreas: effects on islet blood flow and insulin
[45] Bao B, Jiang J, Yanase T, Nishi Y and Morgan J R 2011 secretion in rats Diabetologia 41 127–33
Connexon-mediated cell adhesion drives microtissue self- [55] Mattsson G, Jansson L and Carlsson P O 2002 Decreased
assembly FASEB J. 25 255–64 vascular density in mouse pancreatic islets after
[46] Benavides O M, Brooks A R, Cho S K, Petsche Connell J, transplantation Diabetes 51 1362–6
Ruano R and Jacot J G 2015 In situ vascularization of injectable [56] Carlsson P, Palm F, Andersson A and Liss P 2001 Markedly
fibrin/poly(ethylene glycol) hydrogels by human amniotic decreased oxygen tension in transplanted rat pancreatic islets
fluid-derived stem cells J. Biomed. Mater. Res. A 1–9 irrespective of the implatation site Cell 50 489–95
[47] Lafleur M A, Handsley M M, Knäuper V, Murphy G and
Edwards D R 2002 Endothelial tubulogenesis within fibrin gels

15

Das könnte Ihnen auch gefallen